US20020155491A1 - Arrays for detecting nucleic acids - Google Patents

Arrays for detecting nucleic acids Download PDF

Info

Publication number
US20020155491A1
US20020155491A1 US10/125,428 US12542802A US2002155491A1 US 20020155491 A1 US20020155491 A1 US 20020155491A1 US 12542802 A US12542802 A US 12542802A US 2002155491 A1 US2002155491 A1 US 2002155491A1
Authority
US
United States
Prior art keywords
sequence
sequences
substrate
reagents
target
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/125,428
Inventor
Stephen Fodor
Dennis Solas
William Dower
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.)
Affymetrix Inc
Original Assignee
Affymetrix 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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=24500711&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20020155491(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority claimed from US08/670,118 external-priority patent/US5800992A/en
Application filed by Affymetrix Inc filed Critical Affymetrix Inc
Priority to US10/125,428 priority Critical patent/US20020155491A1/en
Publication of US20020155491A1 publication Critical patent/US20020155491A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • GPHYSICS
    • G03PHOTOGRAPHY; CINEMATOGRAPHY; ANALOGOUS TECHNIQUES USING WAVES OTHER THAN OPTICAL WAVES; ELECTROGRAPHY; HOLOGRAPHY
    • G03FPHOTOMECHANICAL PRODUCTION OF TEXTURED OR PATTERNED SURFACES, e.g. FOR PRINTING, FOR PROCESSING OF SEMICONDUCTOR DEVICES; MATERIALS THEREFOR; ORIGINALS THEREFOR; APPARATUS SPECIALLY ADAPTED THEREFOR
    • G03F7/00Photomechanical, e.g. photolithographic, production of textured or patterned surfaces, e.g. printing surfaces; Materials therefor, e.g. comprising photoresists; Apparatus specially adapted therefor
    • 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
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y10/00Nanotechnology for information processing, storage or transmission, e.g. quantum computing or single electron logic
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y30/00Nanotechnology for materials or surface science, e.g. nanocomposites
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/06Pyrimidine radicals
    • C07H19/10Pyrimidine radicals with the saccharide radical esterified by phosphoric or polyphosphoric acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H21/00Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/04General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length on carriers
    • C07K1/042General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length on carriers characterised by the nature of the carrier
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/04General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length on carriers
    • C07K1/045General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length on carriers using devices to improve synthesis, e.g. reactors, special vessels
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K17/00Carrier-bound or immobilised peptides; Preparation thereof
    • C07K17/02Peptides being immobilised on, or in, an organic carrier
    • C07K17/06Peptides being immobilised on, or in, an organic carrier attached to the carrier via a bridging agent
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K17/00Carrier-bound or immobilised peptides; Preparation thereof
    • C07K17/14Peptides being immobilised on, or in, an inorganic carrier
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6804Nucleic acid analysis using immunogens
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6809Methods for determination or identification of nucleic acids involving differential detection
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6813Hybridisation assays
    • C12Q1/6816Hybridisation assays characterised by the detection means
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6813Hybridisation assays
    • C12Q1/6827Hybridisation assays for detection of mutation or polymorphism
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6813Hybridisation assays
    • C12Q1/6834Enzymatic or biochemical coupling of nucleic acids to a solid phase
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6813Hybridisation assays
    • C12Q1/6834Enzymatic or biochemical coupling of nucleic acids to a solid phase
    • C12Q1/6837Enzymatic or biochemical coupling of nucleic acids to a solid phase using probe arrays or probe chips
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6869Methods for sequencing
    • C12Q1/6874Methods for sequencing involving nucleic acid arrays, e.g. sequencing by hybridisation
    • GPHYSICS
    • G03PHOTOGRAPHY; CINEMATOGRAPHY; ANALOGOUS TECHNIQUES USING WAVES OTHER THAN OPTICAL WAVES; ELECTROGRAPHY; HOLOGRAPHY
    • G03FPHOTOMECHANICAL PRODUCTION OF TEXTURED OR PATTERNED SURFACES, e.g. FOR PRINTING, FOR PROCESSING OF SEMICONDUCTOR DEVICES; MATERIALS THEREFOR; ORIGINALS THEREFOR; APPARATUS SPECIALLY ADAPTED THEREFOR
    • G03F7/00Photomechanical, e.g. photolithographic, production of textured or patterned surfaces, e.g. printing surfaces; Materials therefor, e.g. comprising photoresists; Apparatus specially adapted therefor
    • G03F7/26Processing photosensitive materials; Apparatus therefor
    • G03F7/265Selective reaction with inorganic or organometallic reagents after image-wise exposure, e.g. silylation
    • GPHYSICS
    • G03PHOTOGRAPHY; CINEMATOGRAPHY; ANALOGOUS TECHNIQUES USING WAVES OTHER THAN OPTICAL WAVES; ELECTROGRAPHY; HOLOGRAPHY
    • G03FPHOTOMECHANICAL PRODUCTION OF TEXTURED OR PATTERNED SURFACES, e.g. FOR PRINTING, FOR PROCESSING OF SEMICONDUCTOR DEVICES; MATERIALS THEREFOR; ORIGINALS THEREFOR; APPARATUS SPECIALLY ADAPTED THEREFOR
    • G03F7/00Photomechanical, e.g. photolithographic, production of textured or patterned surfaces, e.g. printing surfaces; Materials therefor, e.g. comprising photoresists; Apparatus specially adapted therefor
    • G03F7/26Processing photosensitive materials; Apparatus therefor
    • G03F7/38Treatment before imagewise removal, e.g. prebaking
    • GPHYSICS
    • G11INFORMATION STORAGE
    • G11CSTATIC STORES
    • G11C13/00Digital stores characterised by the use of storage elements not covered by groups G11C11/00, G11C23/00, or G11C25/00
    • G11C13/0002Digital stores characterised by the use of storage elements not covered by groups G11C11/00, G11C23/00, or G11C25/00 using resistive RAM [RRAM] elements
    • G11C13/0009RRAM elements whose operation depends upon chemical change
    • G11C13/0014RRAM elements whose operation depends upon chemical change comprising cells based on organic memory material
    • GPHYSICS
    • G11INFORMATION STORAGE
    • G11CSTATIC STORES
    • G11C13/00Digital stores characterised by the use of storage elements not covered by groups G11C11/00, G11C23/00, or G11C25/00
    • G11C13/0002Digital stores characterised by the use of storage elements not covered by groups G11C11/00, G11C23/00, or G11C25/00 using resistive RAM [RRAM] elements
    • G11C13/0009RRAM elements whose operation depends upon chemical change
    • G11C13/0014RRAM elements whose operation depends upon chemical change comprising cells based on organic memory material
    • G11C13/0019RRAM elements whose operation depends upon chemical change comprising cells based on organic memory material comprising bio-molecules
    • 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
    • B01J2219/00351Means for dispensing and evacuation of reagents
    • B01J2219/00427Means for dispensing and evacuation of reagents using masks
    • B01J2219/00432Photolithographic masks
    • 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
    • B01J2219/00427Means for dispensing and evacuation of reagents using masks
    • B01J2219/00434Liquid crystal masks
    • 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
    • B01J2219/00436Maskless 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
    • B01J2219/00277Apparatus
    • B01J2219/00457Dispensing or evacuation of the solid phase support
    • B01J2219/00459Beads
    • 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
    • B01J2219/00457Dispensing or evacuation of the solid phase support
    • B01J2219/00459Beads
    • B01J2219/00468Beads by manipulation of individual beads
    • 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
    • B01J2219/00457Dispensing or evacuation of the solid phase support
    • B01J2219/00475Sheets
    • 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
    • B01J2219/005Beads
    • 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
    • B01J2219/00527Sheets
    • 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
    • B01J2219/00527Sheets
    • B01J2219/00529DNA chips
    • 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
    • B01J2219/00527Sheets
    • B01J2219/00531Sheets essentially square
    • 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
    • B01J2219/0059Sequential 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
    • B01J2219/00608DNA chips
    • 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
    • B01J2219/00623Immobilisation 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
    • B01J2219/00623Immobilisation or binding
    • B01J2219/0063Other, e.g. van der Waals forces, hydrogen bonding
    • 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
    • B01J2219/00639Making arrays on substantially continuous surfaces the compounds being trapped in or bound to a porous medium
    • B01J2219/00641Making arrays on substantially continuous surfaces the compounds being trapped in or bound to a porous medium the porous medium being continuous, e.g. porous oxide substrates
    • 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
    • B01J2219/00646Making arrays on substantially continuous surfaces the compounds being bound to beads immobilised on the solid supports
    • B01J2219/00648Making arrays on substantially continuous surfaces the compounds being bound to beads immobilised on the solid supports by the use of solid beads
    • 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
    • B01J2219/00686Automatic
    • B01J2219/00689Automatic using computers
    • 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
    • B01J2219/00695Synthesis control routines, e.g. using computer programs
    • 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
    • B01J2219/00709Type of synthesis
    • B01J2219/00711Light-directed synthesis
    • 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
    • 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
    • B01J2219/00725Peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/11Compounds covalently bound to a solid support
    • 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
    • C40B40/00Libraries per se, e.g. arrays, mixtures
    • C40B40/04Libraries containing only organic compounds
    • C40B40/10Libraries containing peptides or polypeptides, 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
    • G01N15/1433

Definitions

  • the present invention relates to the sequencing, fingerprinting, and mapping of polymers, particularly biological polymers.
  • the inventions may be applied, for example, in the sequencing, fingerprinting, or mapping of nucleic acids, polypeptides, oligosaccharides, and synthetic polymers.
  • Genetic information is critical in continuation of life processes. Life is substantially informationally based and its genetic content controls the growth and reproduction of the organism and its complements. Polypeptides, which are critical features of all living systems, are encoded by the genetic material of the cell. In particular, the properties of enzymes, functional proteins, and structural proteins are determined by the sequence of amino acids which make them up. As structure and function are integrally related, many biological functions may be explained by elucidating the underlying structural features which provide those functions. For this reason, it has become very important to determine the genetic sequences of nucleotides which encode the enzymes, structural proteins, and other effectors of biological functions. In addition to segments of nucleotides which encode polypeptides, there are many nucleotide sequences which are involved in control and regulation of gene expression.
  • the human genome project is directed toward determining the complete sequence of the genome of the human organism. Although such a sequence would not correspond to the sequence of any specific individual, it would provide significant information as to the general organization and specific sequences contained within segments from particular individuals. It would also provide mapping information which is very useful for further detailed studies. However, the need for highly rapid, accurate, and inexpensive sequencing technology is nowhere more apparent than in a demanding sequencing project such as this. To complete the sequencing of a human genome would require the determination of approximately 3 ⁇ 10 9 , or 3 billion base pairs.
  • the procedures typically used today for sequencing include the Sanger dideoxy method, see, e.g., Sanger et al. (1977) Proc. Natl. Acad. Sci. USA, 74:5463-5467, or the Maxam and Gilbert method, see, e.g., Maxam et al., (1980) Methods in Enzymology, 65:499-559.
  • the Sanger method utilizes enzymatic elongation procedures with chain terminating nucleotides.
  • the Maxam and Gilbert method uses chemical reactions exhibiting specificity of reaction to generate nucleotide specific cleavages. Both methods require a practitioner to perform a large number of complex manual manipulations.
  • the present invention provides improved methods useful for de novo sequencing of an unknown polymer sequence, for verification of known sequences, for fingerprinting polymers, and for mapping homologous segments within a sequence. By reducing the number of manual manipulations required and automating most of the steps, the speed, accuracy, and reliability of these procedures are greatly enhanced.
  • a substrate having a matrix of positionally defined regions with attached reagents exhibiting known recognition specificity can be used for the sequence analysis of a polymer. Although most directly applicable to sequencing, the present invention is also applicable to fingerprinting, mapping, and general screening of specific interactions.
  • the VLSIPSTM Technology (Very Large Scale Immobilized Polymer Synthesis) substrates will be applied to evaluating other polymers, e.g., carbohydrates, polypeptides, hydrocarbon synthetic polymers, and the like.
  • the sequence specific reagents will usually be antibodies specific for a particular subunit sequence.
  • the invention provides an ordered method for forming a plurality of polymer sequences by sequential addition of reagents comprising the step of serially protecting and deprotecting portions of the plurality of polymer sequences for addition of other portions of the polymer sequences using a binary synthesis strategy.
  • the present invention also provides a means to automate sequencing manipulations.
  • the automation of the substrate production method and of the scan and analysis steps minimizes the need for human intervention. This simplifies the tasks and promotes reproducibility.
  • the present invention provides a composition comprising a plurality of positionally distinguishable sequence specific reagents attached to a solid substrate, which reagents are capable of specifically binding to a predetermined subunit sequence of a preselected multi-subunit length having at least three subunits, said reagents representing substantially all possible sequences of said preselected length.
  • the subunit sequence is a polynucleotide or a polypeptide, in others the preselected multi-subunit length is five subunits and the subunit sequence is a polynucleotide sequence.
  • the specific reagent is an oligonucleotide of at least about five nucleotides.
  • the specific reagent is a monoclonal antibody.
  • the specific reagents are all attached to a single solid substrate, and the reagents comprise about 3000 different sequences.
  • the reagents represents at least about 25% of the possible subsequences of said preselected length.
  • the reagents are localized in regions of the substrate having a density of at least 25 regions per square centimeter, and often the substrate has a surface area of less than about 4 square centimeters.
  • the present invention also provides methods for analyzing a sequence of a polynucleotide or a polypeptide, said method comprising the step of:
  • the present invention also provides methods for sequencing a segment of a polynucleotide comprising the steps of:
  • a substrate comprising a plurality of chemically synthesized and positionally distinguishable oligonucleotides capable of recognizing defined oligonucleotide sequences
  • the segment is substantially the entire length of said polynucleotide.
  • the invention also provides methods for sequencing a polymer, said method comprising the steps of:
  • the substrates are beads.
  • the plurality of reagents comprise substantially all possible subsequences of said preselected length found in said target.
  • the solid phase substrate is a single substrate having attached thereto reagents recognizing substantially all possible subsequences of preselected length found in said target.
  • the method further comprises the step of analyzing a plurality of said recognized subsequences to assemble a sequence of said target polymer.
  • at least some of the plurality of substrates have one subsequence specific reagent attached thereto, and the substrates are coded to indicate the sequence specificity of said reagent.
  • the present invention also embraces a method of using a fluorescent nucleotide to detect interactions with oligonucleotide probes of known sequence, said method comprising:
  • a method of mapping a plurality of sequences relative to one another comprising:
  • sequence specific probes are oligonucleotides, applicable to where the target sequences are nucleic acid sequences.
  • the steps of the sequencing process comprise:
  • the enablement of the sequencing process by hybridization is based in large part upon the ability to synthesize a large number (e.g., to virtually saturate) of the possible overlapping sequence segments and distinguishing those probes which hybridize with fidelity from those which have mismatched bases, and to analyze a highly complex pattern of hybridization results to determine the overlap regions.
  • the detecting of the positions which bind the target sequence would typically be through a fluorescent label on the target.
  • a fluorescent label is probably most convenient, other sorts of labels, e.g., radioactive, enzyme linked, optically detectable, or spectroscopic labels may be used.
  • the oligonucleotide probes are positionally defined, the location of the hybridized duplex will directly translate to the sequences which hybridize.
  • analysis of the positions provides a collection of subsequences found within the target sequence. These subsequences are matched with respect to their overlaps so as to assemble an intact target sequence.
  • FIG. 1 illustrates a flow chart for sequence, fingerprint, or mapping analysis.
  • FIG. 2A-M illustrates the process of a VLSIPSTM Technology trinucleotide synthesis.
  • the present invention relies in part on the ability to synthesize or attach specific recognition reagents at known locations on a substrate, typically a single substrate.
  • the present invention provides the ability to prepare a substrate having a very high density matrix pattern of positionally defined specific recognition reagents.
  • the reagents are capable of interacting with their specific targets while attached to the substrate, e.g., solid phase interactions, and by appropriate labeling of these targets, the sites of the interactions between the target and the specific reagents may be derived. Because the reagents are positionally defined, the sites of the interactions will define the specificity of each interaction.
  • a map of the patterns of interactions with specific reagents on the substrate is convertible into information on the specific interactions taking place, e.g., the recognized features.
  • this system allows the determination of the combination of specific interactions which exist on the target molecule.
  • the features may actually be the subunit sequence of the target molecule, and a given target sequence may be uniquely defined by its combination of features.
  • the methodology is applicable to sequencing polynucleotides.
  • the specific sequence recognition reagents will typically be oligonucleotide probes which hybridize with specificity to subsequences found on the target sequence. A sufficiently large number of those probes allows the fingerprinting of a target polynucleotide or the relative mapping of a collection of target polynucleotides, as described in greater detail below.
  • polynucleotide sequence analysis is a preferred embodiment, for which the specific reagents are most easily accessible
  • the invention is also applicable to analysis of other polymers, including polypeptides, carbohydrates, and synthetic polymers, including ⁇ -, ⁇ -, and ⁇ -amino acids, polyurethanes, polyesters, polycarbonates, polyureas, polyamides, polyethyleneimines, polyarylene sulfides, polysiloxanes, polyimides, polyacetates, and mixed polymers.
  • Various optical isomers e.g., various D- and L- forms of the monomers, may be used.
  • Sequence analysis will take the form of complete sequence determination, to the level of the sequence of individual subunits along the entire length of the target sequence. Sequence analysis also takes the form of sequence homology, e.g., less than absolute subunit resolution, where “similarity” in the sequence will be detectable, or the form of selective sequences of homology interspersed at specific or irregular locations.
  • the sequence is determinable at selective resolution or at particular locations.
  • the hybridization method will be useful as a means for identification, e.g., a “fingerprint”, much like a Southern hybridization method is used. It is also useful to map particular target sequences.
  • the invention is enabled by the development of technology to prepare substrates on which specific reagents may be either positionally attached or synthesized.
  • VLSIPSTM very large scale immobilized polymer synthesis
  • These reagents specifically recognize subsequences in a target polymer and bind thereto, producing a map of positionally defined regions of interaction.
  • map positions are convertible into actual features recognized, and thus would be present in the target molecule of interest.
  • sequence specific recognition reagents will often be oligonucleotides which hybridize with fidelity and discrimination to the target sequence.
  • monoclonal or polyclonal antibodies having high sequence specificity will often be used.
  • the VLSIPS technology allows the production of a substrate with a high density matrix of positionally mapped regions with specific recognition reagents attached at each distinct region.
  • protective groups which can be positionally removed, or added, the regions can be activated or deactivated for addition of particular reagents or compounds. Details of the protection are described below and in related Pirrung et al. (1992) U.S. Pat. No. 5,143,854.
  • photosensitive protecting agents will be used and the regions of activation or deactivation may be controlled by electro-optical and optical methods, similar to many of the processes used in semiconductor wafer and chip fabrication.
  • VLSIPS substrate is synthesized having positionally defined oligonucleotide probes. See Pirrung et al. (1992) U.S. Pat. No. 5,143,854; and U.S. Ser. No. 07/624,120, now abandoned.
  • the VLSIPS apparatus allows for the stepwise synthesis of polymers according to a positionally defined matrix pattern.
  • Each oligonucleotide probe will be synthesized at known and defined positional locations on the substrate. This forms a matrix pattern of known relationship between position and specificity of interaction.
  • the VLSIPS technology allows the production of a very large number of different oligonucleotide probes to be simultaneously and automatically synthesized including numbers in excess of about 10 2 , 10 3 , 10 4 , 10 5 , 10 6 , or even more, and at densities of at least about 10 2 , 10 3 /cm 2 , 10 4 /cm 2 , 10 5 /cm 2 and up to 10 6 /cm 2 or more.
  • This application discloses methods for synthesizing polymers on a silicon or other suitably derivatized substrate, methods and chemistry for synthesizing specific types of biological polymers on those substrates, apparatus for scanning and detecting whether interaction has occurred at specific locations on the substrate, and various other technologies related to the use of a high density very large scale immobilized polymer substrate.
  • sequencing, fingerprinting, and mapping applications are discussed herein in detail, though related technologies are described in simultaneously filed applications U.S. Ser. No. 07/624,120, now abandoned; and U.S. Ser. No. 07/517,659; Dower et al. (1995) U.S. Pat. No. 5,427,908, each of which is hereby incorporated herein by reference.
  • antibody probes will be generated which specifically recognize particular subsequences found on a polymer.
  • Antibodies would be generated which are specific for recognizing a three contiguous amino acid sequence, and monoclonal antibodies may be preferred. optimally, these antibodies would not recognize any sequences other than the specific three amino acid stretch desired and the binding affinity should be insensitive to flanking or remote sequences found on a target molecule.
  • antibodies specific for particular carbohydrate linkages or sequences will be generated.
  • a similar approach could be used for preparing specific reagents which recognize other polymer subunit sequences. These reagents would typically be site specifically localized to a substrate matrix pattern where the regions are closely packed.
  • reagents could be individually attached at specific sites on the substrate in a matrix by an automated procedure where the regions are positionally targeted by some other specific mechanism, e.g., one which would allow the entire collection of reagents to be attached to the substrate in a single reaction.
  • Each reagent could be separately attached to a specific oligonucleotide sequence by an automated procedure. This would produce a collection of reagents where, e.g., each monoclonal antibody would have a unique oligonucleotide sequence attached to it.
  • each monoclonal antibody would specifically be bound only at that site on the substrate where the complementary oligonucleotide has been synthesized.
  • a crosslinking step would fix the reagent to the substrate. See, e.g., Dattagupta et al. (1985) U.S. Pat. No. 4,542,102 and (1987) U.S. Pat. No. 4,713,326; and Chatterjee, M. et al. (1990) J. Am. Chem. Soc. 112:6397-6399, which are hereby incorporated herein by reference.
  • This allows a high density positionally specific collection of specific recognition reagents, e.g., monoclonal antibodies, to be immobilized to a solid substrate using an automated system.
  • the regions which define particular reagents will usually be generated by selective protecting groups which may be activated or deactivated.
  • the protecting group will be bound to a monomer subunit or spatial region, and can be spatially affected by an activator, such as electromagnetic radiation.
  • an activator such as electromagnetic radiation.
  • protective groups with utility herein include nitroveratryl oxycarbonyl (NVOC), nitrobenzyl oxycarbony (NBOC), dimethyl dimethoxy benzyloxy carbonyl, 5-bromo-7-nitroindolinyl, O-hydroxy- ⁇ -methyl cinnamoyl, and 2-oxymethylene anthraquinone.
  • activators include ion beams, electric fields, magnetic fields, electron beams, x-ray, and other forms of electromagnetic radiation.
  • the binary synthesis strategy refers to an ordered strategy for parallel synthesis of diverse polymer sequences by sequential addition of reagents which may be represented by a reactant matrix, and a switch matrix, the product of which is a product matrix.
  • a reactant matrix is a 1 ⁇ n matrix of the building blocks to be added.
  • the switch matrix is all or a subset of the binary numbers from 1 to n arranged in columns.
  • a binary strategy is one in which at least two successive steps illuminate half of a region of interest on the substrate.
  • binary synthesis refers to a synthesis strategy which also factors a previous addition step.
  • a strategy in which a switch matrix for a masking strategy halves regions that were previously illuminated, illuminating about half of the previously illuminated region and protecting the remaining half (while also protecting about half of previously protected regions and illuminating about half of previously protected regions).
  • binary rounds may be interspersed with non-binary rounds and that only a portion of a substrate may be subjected to a binary scheme, but will still be considered to be a binary masking scheme within the definition herein.
  • a binary “masking” strategy is a binary synthesis which uses light to remove protective groups from materials for addition of other materials such as nucleotides or amino acids.
  • this procedure provides a simplified and highly efficient method for saturating all possible sequences of a defined length polymer.
  • This masking strategy is also particularly useful in producing all possible oligonucleotide sequence probes of a given length.
  • the technology provided by the present invention has very broad applications. Although described specifically for polynucleotide sequences, similar sequencing, fingerprinting, mapping, and screening procedures can be applied to polypeptide, carbohydrate, or other polymers.
  • the present invention may be used to completely sequence a given target sequence to subunit resolution. This may be for de novo sequencing, or may be used in conjunction with a second sequencing procedure to provide independent verification. See, e.g., (1988) Science 242:1245. For example, a large polynucleotide sequence defined by either the Maxam and Gilbert technique or by the Sanger technique may be verified by using the present invention.
  • a polynucleotide sequence can be fingerprinted. Fingerprinting is a less detailed sequence analysis which usually involves the characterization of a sequence by a combination of defined features. Sequence fingerprinting is particularly useful because the repertoire of possible features which can be tested is virtually infinite. Moreover, the stringency of matching is also variable depending upon the application. A Southern Blot analysis may be characterized as a means of simple fingerprint analysis.
  • Fingerprinting analysis may be performed to the resolution of specific nucleotides, or may be used to determine homologies, most commonly for large segments.
  • an array of oligonucleotide probes of virtually any workable size may be positionally localized on a matrix and used to probe a sequence for either absolute complementary matching, or homology to the desired level of stringency using selected hybridization conditions.
  • the present invention provides means for mapping analysis of a target sequence or sequences. Mapping will usually involve the sequential ordering of a plurality of various sequences, or may involve the localization of a particular sequence within a plurality of sequences. This may be achieved by immobilizing particular large segments onto the matrix and probing with a shorter sequence to determine which of the large sequences contain that smaller sequence. Alternatively, relatively shorter probes of known or random sequence may be immobilized to the matrix and a map of various different target sequences may be determined from overlaps. Principles of such an approach are described in some detail by Evans et al. (1989) “Physical Mapping of Complex Genomes by Cosmid Multiplex Analysis,” Proc. Natl. Acad. Sci.
  • Fingerprinting analysis also provides a means of identification.
  • fingerprinting can ensure personal identification for other reasons. For example, it may be useful for identification of bodies in tragedies such as fire, flood, and vehicle crashes. In other cases the identification may be useful in identification of persons suffering from amnesia, or of missing persons.
  • Other forensics applications include establishing the identity of a person, e.g., military identification “dog tags”, or may be used in identifying the source of particular biological samples. Fingerprinting technology is described, e.g., in Carrano, et al.
  • the fingerprinting analysis may be used to perform various types of genetic screening.
  • a single substrate may be generated with a plurality of screening probes, allowing for the simultaneous genetic screening for a large number of genetic markers.
  • prenatal or diagnostic screening can be simplified, economized, and made more generally accessible.
  • the present invention also provides means for determining specificity of interaction with particular sequences. Many of these applications were described in Ser. No. 07/362,901, now abandoned, Pirrung et al. (1992) U.S. Pat. No. 5,143,854; Ser. Nos. 07/435,316, and 07/612,671.
  • Suitable labels include radionucleotides, enzymes, substrates, cofactors, inhibitors, magnetic particles, heavy metal atoms, and particularly fluorescers, chemiluminescers, and spectroscopic labels.
  • Patents teaching the use of such labels include U.S. Pat. Nos. 3,817,837; 3,850,752; 3,939,350; 3,996,345; 4,277,437; 4,275,149; and 4,366,241.
  • the detection system best adapted for high resolution and high sensitivity detection may be selected.
  • an optically detectable system e.g., fluorescence or chemiluminescence would be preferred.
  • other detection systems may be adapted to the purpose, e.g., electron microscopy, scanning electron microscopy (SEM), scanning tunneling electron microscopy (STEM), infrared microscopy, atomic force microscopy (AFM), electrical condutance, and image plate transfer.
  • the initial data resulting from the detection system is an array of data indicative of fluorescent intensity versus location on the substrate.
  • the data are typically taken over regions substantially smaller than the area in which synthesis of a given polymer has taken place.
  • the data may be taken over regions having dimensions of 5 microns by 5 microns.
  • the regions over which florescence data are taken across the substrate are less than about 1 ⁇ 2 the area of the regions in which individual polymers are synthesized, preferably less than ⁇ fraction (1/10) ⁇ the area in which a single polymer is synthesized, and most preferably less than ⁇ fraction (1/100) ⁇ the area in which a single polymer is synthesized.
  • a plot of number of pixels versus intensity for a scan should bear a rough resemblance to a bell curve, but spurious data are observed, particularly at higher intensities. Since it is desirable to use an average of fluorescent intensity over a given synthesis region in determining relative binding affinity, these spurious data will tend to undesirably skew the data.
  • the data are corrected for removal of these spurious data points, and an average of the data points is thereafter utilized in determining relative binding efficiency.
  • the data are fitted to a base curve and statistical measures are used to remove spurious data.
  • the principle of the hybridization sequencing procedure is based, in part, upon the ability to determine overlaps of short segments.
  • the VLSIPS technology provides the ability to generate reagents which will saturate the possible short subsequence recognition possibilities.
  • the principle is most easily illustrated by using a binary sequence, such as a sequence of zeros and ones. Once having illustrated the application to a binary alphabet, the principle may easily be understood to encompass three letter, four letter, five or more letter, even 20 letter alphabets.
  • a theoretical treatment of analysis of subsequence information to reconstruction of a target sequence is provided, e.e., in Lysov, Yu., et al. (1988) Doklady Akademi. Nauk.
  • the reagents for recognizing the subsequences will usually be specific for recognizing a particular polymer subsequence anywhere within a target polymer. It is preferable that conditions may be devised which allow absolute discrimination between high fidelity matching and very low levels of mismatching.
  • the reagent interaction will preferably exhibit no sensitivity to flanking sequences, to the subsequence position within the target, or to any other remote structure within the sequence.
  • the specific reagents can be oligonucleotide probes; for polypeptides and carbohydrates, antibodies will be useful reagents. Antibody reagents should also be useful for other types of polymers.
  • a VLSIPSTM Technology substrate could be constructed, as discussed elsewhere, which would have reagents attached in a defined matrix pattern which specifically recognize each of the possible five digit sequences of ones and zeros.
  • the first five digit sequence contained in the target sequence is 10100.
  • the second is 01001
  • the third is 10011
  • the fourth is 00111
  • the fifth 01110
  • the sixth is 11100.
  • the VLSIPSTM substrate would have a matrix pattern of positionally attached reagents which recognize each of the different 5-mer subsequences. Those reagents which recognize each of the 6 contained 5-mers will bind the target, and a label allows the positional determination of where the sequence specific interaction has occurred. By correlation of the position in the matrix pattern, the corresponding bound subsequences can be determined.
  • This 192 is derived from the observation that with the subsequence 10100 at the far left of the sequence, in positions 1-5, there are only 32 possible sequences. Likewise, for that particular subsequence in positions 2-6, 3-7, 4-8, 5-9, and 6-10. So, to sum up all of the sequences that could contain 10100, there are 32 for each position and 6 positions for a total of about 192 possible sequences. However, some of these 10 digit sequences will have been counted twice. Thus, by virtue of containing the 10100 subsequence, the number of possible 10-mer sequences has been decreased from 1024 sequences to less than about 192 sequences.
  • next 5-mer must be of the sequence 0111W where W must be 0 or 1. Again, looking up at the fragments produced, we see that the target sequence contains a 01110 subsequence, and W is a 0. Thus, our sequence to this point is 101001110. We know that the last 5-mer must be either 11100 or 11101. Looking above, we see that it is 11100 and that must be the last of our sequence. Thus, we have determined that our sequence must have been 1010011100.
  • the sequence analysis can start with any known positive probe subsequence.
  • the determination may be performed by moving linearly along the sequence checking the known sequence with a limited number of next positions.
  • the sequence may be determined, besides by scanning all possible oligonucleotide probe positions, by specifically looking only where the next possible positions would be. This may increase the complexity of the scanning but may provide a longer time span dedicated towards scanning and detecting specific positions of interest relative to other sequence possibilities.
  • the scanning apparatus could be set up to work its way along a sequence from a given contained oligonucleotide to only look at those positions on the substrate which are expected to have a positive signal.
  • the number of possible sequences of a given length increases very quickly with the length of that sequence.
  • a 10-mer of zeros and ones has 1024 possibilities
  • a 12-mer has 4096.
  • a 20-mer has over a million possibilities
  • a 30-mer has over a billion.
  • a given 30-mer has, at most, 26 different internal 5-mer sequences.
  • a 30 character target sequence having over a million possible sequences can be substantially defined by only 26 different 5-mers.
  • the probe oligonucleotides will preferably, but need not necessarily, be of identical length, and that the probe sequences need not necessarily be contiguous in that the overlapping subsequences need not differ by only a single subunit.
  • each position of the matrix pattern need not be homogeneous, but may actually contain a plurality of probes of known sequence.
  • all of the possible subsequence specifications would be preferred, a less than full set of sequences specifications could be used.
  • a substantial fraction will preferably be at least about 70%, it may be less than that. About 20% would be preferred, more preferably at least about 30% would be desired. Higher percentages would be especially preferred.
  • a four letter alphabet may be conceptualized in at least two different ways from the two letter alphabet. One way is to consider the four possible values at each position and to analogize in a similar fashion to the binary example each of the overlaps. A second way is to group the binary digits into groups.
  • the binary system can be looked at in pairs of digits.
  • the pairs would be 00, 01, 10, and 11.
  • the earlier used sequence 1010011100 is looked at as 10, 10, 01, 11, 00.
  • the first character of two digits is selected from the possible universe of the four representations 00, 01, 10, and 11.
  • a probe would be in an even number of digits, e.g., not five digits, but, three pairs of digits or six digits.
  • a similar comparison is performed and the possible overlaps determined.
  • the 3-pair subsequences are:
  • This reconstruction process may be applied to polymers of virtually any number of possible characters in the alphabet, and for virtually any length sequence to be sequenced, though limitations, as discussed below, will limit its efficiency at various extremes of length. It will be recognized that the theory can be applied to a large diversity of systems where sequence is important.
  • the method could be applied to sequencing of a polypeptide.
  • a polypeptide can have any of twenty natural amino acid possibilities at each position.
  • a twenty letter alphabet is amenable to sequencing by this method so long as reagents exist for recognizing shorter subsequences therein.
  • a preferred reagent for achieving that goal would be a set of monoclonal antibodies each of which recognizes a specific three contiguous amino acid subsequence.
  • each target sequence which is recognized by the specific reagents need not have homogeneous termini. Thus, fragments of the entire target sequence will also be useful for hybridizing appropriate subsequences. It is, however, preferable that there not be a significant amount of labeled homogeneous contaminating extraneous sequences. This constraint does usually require the purification of the target molecule to be sequenced, but a specific label technique would dispense with a purification requirement if the unlabeled extraneous sequences do not interfere with the labeled sequences.
  • conformational effects of target polypeptide folding may, in certain embodiments, be negligible if the polypeptide is fragmented into sufficiently small peptides, or if the interaction is performed under conditions where conformation, but not specific interaction, is disrupted.
  • the first obvious complication is a problem which arises from an inappropriate length of recognition sequence, which causes problems with the specificity of recognition. For example, if the recognized sequence is too short, every sequence which is utilized will be recognized by every probe sequence. This occurs, e.g., in a binary system where the probes are each of sequences which occur relatively frequently, e.g., a two character probe for the binary system. Each possible two character probe would be expected to appear 1 ⁇ 4 of the time in every single two character position. Thus, the above sequence example would be recognized by each of the 00, 10, 01, and 11. Thus, the sequence information is virtually lost because the resolution is too low and each recognition reagent specifically binds at multiple sites on the target sequence.
  • the number of different probes which bind to a target depends on the relationship between the probe length and the target length. At the extreme of short probe length, the just mentioned problem exists of excessive redundancy and lack of resolution. The lack of stability in recognition will also be a problem with extremely short probes. At the extreme of long probe length, each entire probe sequence is on a different position of a substrate. However, a problem arises from the number of possible sequences, which goes up dramatically with the length of the sequence. Also, the specificity of recognition begins to decrease as,the contribution to binding by any particular subunit may become sufficiently low that the system fails to distinguish the fidelity of recognition.
  • Mismatched hybridization may be a problem with the polynucleotide sequencing applications, though the fingerprinting and mapping applications may not be so strict in their fidelity requirements.
  • a thirty position binary sequence has over a million possible sequences, a number which starts to become unreasonably large in its required number of different sequences, even though the target length is still very short.
  • Preparing a substrate with all sequence possibilities for a long target may be extremely difficult due to the many different oligomers which must be synthesized.
  • the above example illustrates how a long target sequence may be reconstructed with a reasonably small number of shorter subsequences. Since the present day resolution of the regions of the substrate having defined oligomer probes attached to the substrate approaches about 10 microns by 10 microns for resolvable regions, about 10 6 , or 1 million, positions can be placed on a one centimeter square substrate. However, high resolution systems may have particular disadvantages which may be outweighed using the lower density substrate matrix pattern. For this reason, a sufficiently large number of probe sequences can be utilized so that any given target sequence may be determined by hybridization to a relatively small number of probes.
  • a second complication relates to convergence of sequences to a single subsequence. This will occur when a particular subsequence is repeated in the target sequence.
  • This problem can be addressed in at least two different ways. The first, and simpler way, is to separate the repeat sequences onto two different targets. Thus, each single target will not have the repeated sequence and can be analyzed to its end. This solution, however, complicates the analysis by requiring that some means for cutting at a site between the repeats can be located. Typically a careful sequencer would want to have two intermediate cut points so that the intermediate region can also be sequenced in both directions across each of the cut points. This problem is inherent in the hybridization method for sequencing but can be minimized by using a longer known probe sequence so that the frequency of probe repeats is decreased.
  • PCR polymerase chain reaction
  • Probes can be made to hybridize to those known sequences adjacent the repeat sequences, thereby producing new target sequences for analysis. See, e.g., Innis et al. (eds.) (1990) PCR Protocols: A Guide to Methods and Applications, Academic Press; and methods for synthesis of oligonucleotide probes, see, e.g., Gait (1984) Oligonucleotide Synthesis: A Practical Approach, IRL Press, Oxford.
  • the above example is directed towards a polynucleotide embodiment.
  • This application is relatively easily achieved because the specific reagents will typically be complementary oligonucleotides, although in certain embodiments other specific reagents may be desired.
  • the polynucleotide targets will usually be single strand, but may be double or triple stranded in various applications. However, a triple stranded specific interaction might be sometimes desired, or a protein or other specific binding molecule may be utilized.
  • various promoter or DNA sequence specific binding proteins might be used, including, e.g., restriction enzyme binding domains, other binding domains, and antibodies.
  • specific recognition reagents besides oligonucleotides may be utilized.
  • the specific reagents will often be polypeptides. These polypeptides may be protein binding domains from enzymes or other proteins which display specificity for binding. Usually an antibody molecule may be used, and monoclonal antibodies may be particularly desired.
  • Classical methods may be applied for preparing antibodies, see, e.g., Harlow and Lane (1988) Antibodies: A Laboratory Manual Cold Spring Harbor Press, New York; and Goding (1986) Monoclonal Antibodies: Principles and Practice (2d Ed.) Academic Press, San Diego. Other suitable techniques for in vitro exposure of lymphocytes to the antigens or selection of libraries of antibody binding sites are described, e.g., in Huse et al.
  • the antibody specific reagents should be particularly useful for the polypeptide, carbohydrate, and synthetic polymer applications. Individual specific reagents might be generated by an automated process to generate the number of reagents necessary to advantageously use the high density positional matrix pattern. In an alternative approach, a plurality of hybridoma cells may be screened for their ability to bind to a VLSIPS matrix possessing the desired sequences whose binding specificity is desired. Each cell might be individually grown up and its binding specificity determined by VLSIPS apparatus and technology. An alternative strategy would be to expose the same VLSIPS matrix to a polyclonal serum of high titer.
  • each region of the VLSIPS substrate would have attached to it a substantial number of antibody molecules with specificity of binding.
  • the substrate, with non-covalently bound antibodies could be derivatized and the antibodies transferred to an adjacent second substrate in the matrix pattern in which the antibody molecules had attached to the first matrix. If the sensitivity of detection of binding interaction is sufficiently high, such a low efficiency transfer of antibody molecules may produce a sufficiently high signal to be useful for many purposes, including the sequencing applications.
  • capillary forces may be used to transfer the selected reagents to a new matrix, to which the reagents would be positionally attached in the pattern of the recognized sequences.
  • the reagents could be transversely electrophoresed, magnetically transferred, or otherwise transported to a new substrate in their retained positional pattern.
  • oligonucleotides used in polynucleotide sequencing may be produced in at least two different ways.
  • this collection of reagents may be individually positionally attached to a substrate, thereby allowing a batchwise hybridization step.
  • Present technology also would allow the possibility of attaching each and every one of these 10-mers to a separate specific position on a solid matrix. This attachment could be automated in any of a number of ways, particularly through the use of a caged biotin type linking. This would produce a matrix having each of different possible 10-mers.
  • a batchwise hybridization is much preferred because of its reproducibility and simplicity.
  • An automated process of attaching various reagents to positionally defined sites on a substrate is provided in Pirrung et al. (1992) U.S. Pat. No. 5,143,854; Ser. No. 07/624,120, now abandoned; and Barrett et al. (1993) U.S. Pat. No. 5,252,743; each of which is hereby incorporated herein by reference.
  • each oligonucleotide is conveniently synthesized in parallel by sequential synthetic processes on a defined matrix pattern as provided in Pirrung et al. (1992) U.S. Pat. No. 5,143,854; and Ser. No. 07/624,120, now abandoned, which are incorporated herein by reference.
  • the oligonucleotides are synthesized stepwise on a substrate at positionally separate and defined positions.
  • Use of photosensitive blocking reagents allows for defined sequences of synthetic steps over the surface of a matrix pattern.
  • the surface of the substrate can be positioned to generate a desired pattern of regions, each having a defined sequence oligonucleotide synthesized and immobilized thereto.
  • the regions for synthesis may be very small, usually less than about 100 ⁇ m ⁇ 100 ⁇ m, more usually less than about 50 ⁇ m ⁇ 50 ⁇ m.
  • the photolithography technology allows synthetic regions of less than about 10 ⁇ m ⁇ 10 ⁇ m, about 3 ⁇ m ⁇ 3 ⁇ m, or less.
  • the detection also may detect such sized regions, though larger areas are more easily and reliably measured.
  • Region size is sufficiently small to correspond to densities of at least about 5 regions/cm 2 , 20 regions/cm 2 , 50 regions/cm 2 , 100 regions/cm 2 , and greater, including 300 regions/cm 2 , 1000 regions/cm 2 , 3K regions/cm 2 , 10K regions/cm 2 , 30K regions/cm 2 , 100K regions/cm 2 , 300K regions/cm 2 or more, even in excess of one million regions/cm 2 .
  • the strategy for generating a specific pattern may take any of a number of different approaches. These approaches are well described in related application Ser. No. 07/624,120, now abandoned, and include a number of binary masking approaches which will not be exhaustively discussed herein. However, the binary masking and binary synthesis approaches provide a maximum of diversity with a minimum number of actual synthetic steps.
  • oligonucleotides used in sequencing applications will be selected on criteria determined to some extent by the practical limits discussed above. For example, if probes are made as oligonucleotides, there will be 65,536 possible eight nucleotide sequences. If a nine subunit oligonucleotide is selected, there are 262,144 possible permeations of sequences. If a ten-mer oligonucleotide is selected, there are 1,048,576 possible permeations of sequences. As the number gets larger, the required number of positionally defined subunits necessary to saturate the possibilities also increases. With respect to hybridization conditions, the length of the matching necessary to confer stability of the conditions selected can be compensated for. See, e.g., Kanehisa, M. (1984) Nuc. Acids Res. 12:203-213, which is hereby incorporated herein by reference.
  • the VLSIPS technology would typically use a photosensitive protective group on an oligonucleotide.
  • Sample oligonucleotides are shown in FIG. 1.
  • the photoprotective group on the nucleotide molecules may be selected from a wide variety of positive light reactive groups preferably including nitro aromatic compounds such as o-nitro-benzyl derivatives or benzylsulfonyl. See, e.g., Gait (1984) Oligonucleotide Synthesis: A Practical Approach, IRL Press, oxford, which is hereby incorporated herein by reference.
  • 6-nitro-veratryl oxycarbony NVOC
  • 2-nitrobenzyl oxycarbonyl NBOC
  • ⁇ , ⁇ -dimethyl-dimethoxybenzyl oxycarbonyl DEZ
  • Photoremovable protective groups are described in, e.g., Patchornik (1970) J. Amer. Chem. Soc. 92:6333-6335; and Amit et al. (1974) J. Orcanic Chem. 39:192-196; each of which is hereby incorporated herein by reference.
  • a preferred linker for attaching the oligonucleotide to a silicon matrix is illustrated in FIG. 2. A more detailed description is provided below.
  • a photosensitive blocked nucleotide may be attached to specific locations of unblocked prior cycles of attachments on the substrate and can be successively built up to the correct length oligonucleotide probe.
  • each substrate may be simultaneously exposed to a single target sequence where each substrate is a duplicate of one another or where, in combination, multiple substrates together provide the complete or desired subset of possible subsequences.
  • This provides the opportunity to overcome a limitation of the density of positions on a single substrate by using multiple substrates.
  • each probe might be attached to a single bead or substrate and the beads sorted by whether there is a binding interaction. Those beads which do bind might be encoded to indicate the subsequence specificity of reagents attached thereto.
  • the target may be bound to the whole collection of beads and those beads that have appropriate specific reagents on them will bind to the target.
  • a sorting system may be utilized to sort those beads that actually bind the target from those that do not. This may be accomplished by presently available cell sorting devices or a similar apparatus.
  • the encoding scheme may be read off to determine the specificity of the reagent on the bead.
  • An encoding system may include a magnetic system, a shape encoding system, a color encoding system, or a combination of any of these, or any other encoding system.
  • the parameters of polynucleotide sizes of both the probes and target sequences are determined by the applications and other circumstances.
  • the length of the oligonucleotide probes used will depend in part upon the limitations of the VLSIPS technology to provide the number of desired probes. For example, in an absolute sequencing application, it is often useful to have virtually all of the possible oligonucleotides of a given length. As indicated above, there are 65,536 8-mers, 262,144 9-mers, 1,048,576 10-mers, 4,194,304 11-mers, etc. As the length of the oligomer increases the number of different probes which must be synthesized also increases at a rate of a factor of 4 for every additional nucleotide.
  • the length of the probe is selected for a length that will allow the probe to bind with specificity to possible targets.
  • the hybridization conditions are also very important in that they will determine how closely the homology of complementary binding will be detected.
  • a single target may be evaluated at a number of different conditions to determine its spectrum of specificity for binding particular probes. This may find use in a number of other applications besides the polynucleotide sequencing fingerprinting or mapping. For example, it will be desired to determine the spectrum of binding affinities and specificities of cell surface antigens with binding by particular antibodies immobilized on the substrate surface, particularly under different interaction conditions.
  • regions with reagents having differing affinities or levels of specificity may allow such a spectrum to be defined using a single incubation, where various regions, at a given hybridization condition, show the binding affinity.
  • fingerprint probes of various lengths, or with specific defined non-matches may be used.
  • Unnatural nucleotides or nucleotides exhibiting modified specificity of complementary binding are described in greater detail in Macevicz (1990) PCT pub. No. WO 90/04652; and see the section on modified nucleotides in the Sigma Chemical Company catalogue.
  • the label used to detect the target sequences will be determined, in part, by the detection methods being applied. Thus, the labeling method and label used are selected in combination with the actual detecting systems being used.
  • carbohydrate labeling is described, e.g., in Chaplin and Kennedy (1986) Carbohydrate Analysis: A Practical Approach, IRL Press, Oxford. Labeling of other polymers will be performed by methods applicable to them as recognized by a person having ordinary skill in manipulating the corresponding polymer.
  • the target need not actually be labeled if a means for detecting where interaction takes place is available.
  • a means for detecting where interaction takes place may be provided by an intercalating dye which intercalates only into double stranded segments, e.g., where interaction occurs. See, e.g., Sheldon et al. U.S. Pat. No. 4,582,789.
  • the target sequence will be absolutely homogeneous, both with respect to the total sequence and with respect to the ends of each molecule. Homogeneity with respect to sequence is important to avoid ambiguity. It is preferable that the target sequences of interest not be contaminated with a significant amount of labeled contaminating sequences. The extent of allowable contamination will depend on the sensitivity of the detection system and the inherent signal to noise of the system. Homogeneous contamination sequences will be particularly disruptive of the sequencing procedure.
  • the target polynucleotide must have a unique sequence
  • the target molecules need not have identical ends.
  • the homogeneous target molecule preparation may be randomly sheared to increase the numerical number of molecules. Since the total information content remains the same, the shearing results only in a higher number of distinct sequences which may be labeled and bind to the probe. This fragmentation may give a vastly superior signal relative to a preparation of the target molecules having homogeneous ends.
  • the signal for the hybridization is likely to be dependent on the numerical frequency of the target-probe interactions. If a sequence is individually found on a larger number of separate molecules a better signal will result.
  • shearing a homogeneous preparation of the target may often be preferred before the labeling procedure is performed, thereby producing a large number of labeling groups associated with each subsequence.
  • hybridization conditions between probe and target should be selected such that the specific recognition interaction, i.e., hybridization, of the two molecules is both sufficiently specific and sufficiently stable. See, e.g., Hames and Higgins (1985) Nucleic Acid Hybridisation: A practical Approach, IRL Press, Oxford. These conditions will be dependent both on the specific sequence and often on the guanine and cytosine (GC) content of the complementary hybrid strands. The conditions may often be selected to be universally equally stable independent of the specific sequences involved. This typically will make use of a reagent such as an alkylammonium buffer. See, Wood et al.
  • Temperature and salt conditions along with other buffer parameters should be selected such that the kinetics of renaturation should be essentially independent of the specific target subsequence or oligonucleotide probe involved.
  • the hybridization reactions will usually be performed in a single incubation of all the substrate matrices together exposed to the identical same target probe solution under the same conditions.
  • various substrates may be individually treated differently. Different substrates may be produced, each having reagents which bind to target subsequences with substantially identical stabilities and kinetics of hybridization. For example, all of the high GC content probes could be synthesized on a single substrate which is treated accordingly. In this embodiment, the arylammonium buffers could be unnecessary. Each substrate is then treated in a manner such that the collection of substrates show essentially uniform binding and the hybridization data of target binding to the individual substrate matrix is combined with the data from other substrates to derive the necessary subsequence binding information.
  • the hybridization conditions will usually be selected to be sufficiently specific such that the fidelity of base matching will be properly discriminated. Of course, control hybridizations should be included to determine the stringency and kinetics of hybridization.
  • the next step of the sequencing process by hybridization involves labeling of target polynucleotide molecules.
  • a quickly and easily detectable signal is preferred.
  • the VLSIPSTM Technology apparatus is designed to easily detect a fluorescent label, so fluorescent tagging of the target sequence is preferred.
  • Other suitable labels include heavy metal labels, magnetic probes, chromogenic labels (e.g., phosphorescent labels, dyes, and fluorophores) spectroscopic labels, enzyme linked labels, radioactive labels, and labeled binding proteins. Additional labels are described in U.S. Pat. No. 4,366,241, which is incorporated herein by reference.
  • the detection method provides a positional localization of the region where hybridization has taken place. However, the position is correlated with the specific sequence of the probe since the probe has specifically been attached or synthesized at a defined substrate matrix position. Having collected all of the data indicating the subsequences present in the target sequence, this data may be aligned by overlap to reconstruct the entire sequence of the target, as illustrated above.
  • the matrix substrate should be reusable and readily prepared for exposure to a second or subsequent target polynucleotides.
  • the hybrid duplexes are disrupted and the matrix treated in a way which removes all traces of the original target.
  • the matrix may be treated with various detergents or solvents to which the substrate, the oligonucleotide probes, and the linkages to the substrate are inert. This treatment may include an elevated temperature treatment, treatment with organic or inorganic solvents, modifications in pH, and other means for disrupting specific interaction.
  • a second target may actually be applied to the recycled matrix and analyzed as before.
  • sequence recognition reagents will take the form, typically, of proteins exhibiting binding specificity, e.g., enzyme binding sites or antibody binding sites.
  • Enzyme binding sites may be derived from promoter proteins, restriction enzymes, and the like. See, e.g., Stryer, L. (1988) Biochemistry, W. H. Freeman, Palo Alto. Antibodies will typically be produced using standard procedures, see, e.g., Harlow and Lane (1988) Antibodies: A Laboratory Manual, Cold Spring Harbor Press, New York; and Goding (1986) Monoclonal Antibodies: Principles and Practice, (2d Ed.) Academic Press, San Diego.
  • an antigen, or collection of antigens are presented to an immune system.
  • This may take the form of synthesized short polymers produced by the VLSIPS technology, or by the other synthetic means, or from isolation of natural products.
  • antigen for the polypeptides may be made by the VLSIPS technology, by standard peptide synthesis, by isolation of natural proteins with or without degradation to shorter segments, or by expression of a collection of short nucleic acids of random or defined sequences. See, e.g., Tuerk and Gold (1990) Science 249:505-510, for generation of a collection of randomly mutagenized oligonucleotides useful for expression.
  • the antigen or collection is presented to an appropriate immune system, e.g., to a whole animal as in a standard immunization protocol, or to a collection of immune cells or equivalent.
  • an appropriate immune system e.g., to a whole animal as in a standard immunization protocol, or to a collection of immune cells or equivalent.
  • Ward et al. (1989) Nature 341:544-546; and Huse et al. (1989) Science 246:1275-1281 each of which is hereby incorporated herein by reference.
  • Antibodies may be purified having the desired sequence specificities by isolating the cells producing them.
  • a VLSIPS substrate with the desired antigens synthesized thereon may be used to isolate cells with cell surface reagents which recognize the antigens.
  • the VLSIPS substrate may be used as an affinity reagent to select and recover the appropriate cells.
  • Antibodies from those cells may be attached to a substrate using the caged biotin methodology, or by attaching a targeting molecule, e.g., an oligonucleotide.
  • the supernatants from antibody producing cells can be easily assayed using a VLSIPS substrate to identify the cells producing the appropriate antibodies.
  • cells may be isolated, specific antibody molecules which perform the sequence recognition will also be sufficient.
  • populations of antibody with a known specificity can be isolated.
  • Supernatants from a large population of producing cells may be passed over a VLSIPS substrate to bind to the desired antigens attached to the substrate.
  • a sufficient density of antibody molecules are attached, they may be removed by an automated process, preferably as antibody populations exhibiting specificity of binding.
  • a VLSIPS substrate e.g., with a large plurality of fingerprint antigens attached thereto, is used to isolate antibodies from a supernatant of a population of cells producing antibodies to the antigens.
  • the substrate as an affinity reagent, the antibodies will attach to the appropriate positionally defined antigens.
  • the antibodies may be carefully removed therefrom, preferably by an automated system which retains their homogeneous specificities.
  • the isolated antibodies can be attached to a new substrate in a positionally defined matrix pattern.
  • these spatially separated antibodies may be isolated using a specific targeting method for isolation.
  • a linker molecule which attaches to a particular portion of the antibody, preferably away from the binding site, can be attached to the antibodies.
  • Various reagents will be used, including staphylococcus protein A or antibodies which bind to domains remote from the binding site.
  • the antibodies in the population, before affinity purification may be derivatized with an appropriate reagent compatible with new VLSIPS synthesis.
  • a preferred reagent is a nucleotide which can serve as a linker to synthetic VLSIPS steps for synthesizing a specific sequence thereon.
  • each of the antibodies attached to the defined antigen regions can have a defined oligonucleotide synthesized thereon and corresponding in area to the region of the substrate having each antigen attached.
  • These defined oligonucleotides will be useful as targeting reagents to attach those antibodies possessing the same target sequence specificity at defined positions on a new substrate, by virtue of having bound to the antigen region, to a new VLSIPS substrate having the complementary target oligonucleotides positionally located on it.
  • a VLSIPS substrate having the desired antigens attached thereto can be used to generate a second VLSIPS substrate with positionally defined reagents which recognize those antigens.
  • the selected antigens will typically be selected to be those which define particular functionalities or properties, so as to be useful for fingerprinting and other uses. They will also be useful for mapping and sequencing embodiments.
  • sequencing embodiment is very useful in identifying the probes useful in further fingerprinting uses. For example, characteristic features of genetic sequences will be identified as being diagnostic of the entire sequence. However, in most embodiments, longer probe and target will be used, and for which slight mismatching may not need to be resolved.
  • a collection of specific probes may be produced by either of the methods described above in the section on sequencing.
  • Specific oligonucleotide probes of desired lengths may be individually synthesized on a standard oligonucleotide synthesizer. The length of these probes is limited only by the ability of the synthesizer to continue to accurately synthesize a molecule.
  • Oligonucleotides or sequence fragments may also be isolated from natural sources.
  • Biological amplification methods may be coupled with synthetic synthesizing procedures such as, e.g., polymerase chain reaction.
  • the individually isolated probes may be attached to the matrix at defined positions.
  • These probe reagents may be attached by an automated process making use of the caged biotin methodology described in Ser. No. 07/612,671, or using photochemical reagents, see, e.g., Dattagupta et al. (1985) U.S. Pat. No. 4,542,102 and (1987) U.S. Pat. No. 4,713,326.
  • Each individually purified reagent can be attached individually at specific locations on a substrate.
  • the VLSIPS synthesizing technique may be used to synthesize the desired probes at specific positions on a substrate.
  • the probes may be synthesized by successively adding appropriate monomer subunits, e.g., nucleotides, to generate the desired sequences.
  • a relatively short specific oligonucleotide which serves as a targeting reagent for positionally directing the sequence recognition reagent.
  • the sequence specific reagents having a separate additional sequence recognition segment can be directed to target oligonucleotides attached to the substrate.
  • non-natural targeting reagents e.g., unusual nucleotide analogues which pair with other unnatural nucleotide analogues and which do not interfere with natural nucleotide interactions, the natural and non-natural portions can coexist on the same molecule without interfering with their individual functionalities.
  • the two are crosslinked, thereby permanently attaching them to the substrate.
  • Suitable crosslinking reagents are known, see, e.g., Dattagupta et al. (1985) U.S. Pat. No. 4,542,102 and (1987) “Coupling of nucleic acids to solid support by photochemical methods,” U.S. Pat. No. 4,713,326, each of which is hereby incorporated herein by reference.
  • Similar linkages for attachment of proteins to a solid substrate are provided, e.g., in Merrifield (1986) Science 232:341-347, which is hereby incorporated herein by reference.
  • the labeling procedures used in the sequencing embodiments will also be applicable in the fingerprinting embodiments.
  • the fingerprinting embodiments often will involve relatively large target molecules and relatively short oligonucleotide probes, the amount of signal necessary to incorporate into the target sequence may be less critical than in the sequencing applications. For example, a relatively long target with a relatively small number of labels per molecule may be easily amplified or detected because of the relatively large target molecule size.
  • the hybridization conditions used in fingerprinting embodiments will typically be less critical than for the sequencing embodiments. The reason is that the amount of mismatching which may be useful in providing the fingerprinting information would typically be far greater than that necessary in sequencing uses. For example, Southern hybridizations do not typically distinguish between slightly mismatched sequences. Under these circumstances, important and valuable information may be arrived at with less stringent hybridization conditions while providing valuable fingerprinting information. However, since the entire substrate is typically exposed to the target molecule at one time, the binding affinity of the probes should usually be of approximately comparable levels. For this reason, if oligonucleotide probes are being used, their lengths should be approximately comparable and will be selected to hybridize under conditions which are common for most of the probes on the substrate.
  • the target and oligonucleotide probes are of lengths typically greater than about 25 nucleotides. Under appropriate hybridization conditions, e.g., typically higher salt and lower temperature, the probes will hybridize irrespective of imperfect complementarity. In fact, with probes of greater than, e.g., about fifty nucleotides, the difference in stability of different sized probes will be relatively minor.
  • the fingerprinting is merely for probing similarity or homology.
  • the stringency of hybridization can usually be decreased to fairly low levels. See, e.g., Wetmur and Davidson (1968) “Kinetics of Renaturation of DNA,” J. Mol. Biol., 31:349-370; and Kanehisa, M. (1984) Nuc. Acids Res., 12:203-213.
  • Detection methods will be selected which are appropriate for the selected label.
  • the scanning device need not necessarily be digitized or placed into a specific digital database, though such would most likely be done.
  • the analysis in fingerprinting could be photographic.
  • the pattern of hybridizations may be spatially unique and may be compared photographically. In this manner, each sample may have a characteristic pattern of interactions and the likelihood of identical patterns will preferably be such low frequency that the fingerprint pattern indeed becomes a characteristic pattern virtually as unique as an individual's fingertip fingerprint.
  • every individual could be, in theory, uniquely identifiable on the basis of the pattern of hybridizing to the substrate.
  • the VLSIPSTM Technology scanning apparatus may also be useful to generate a digitized version of the fingerprint pattern.
  • the identification pattern can be provided in a linear string of digits.
  • This sequence could also be used for a standardized identification system providing significant useful medical transferability of specific data.
  • the probes used are selected to be of sufficiently high resolution to measure the antigens of the major histo compatibility complex. It might even be possible to provide transplantation matching data in a linear stream of data.
  • the fingerprinting data may provide a condensed version, or summary, of the linear genetic data, or any other information data base.
  • the analysis of the fingerprint will often be much simpler than a total sequence determination.
  • there may be particular types of analysis which will be substantially simplified by a selected group of probes. For example, probes which exhibit particular populational heterogeneity may be selected. In this way, analysis may be simplified and practical utility enhanced merely by careful selection of the specific probes and a careful matrix layout of those probes.
  • the fingerprinting usages may also take advantage of the reusability of the substrate. In this way, the interactions can be disrupted, the substrate treated, and the renewed substrate is equivalent to an unused substrate.
  • the fingerprinting analysis may be applied to other polymers, especially polypeptides, carbohydrates, and other polymers, both organic and inorganic.
  • the fingerprinting method may be used to characterize various samples. For example, a cell or population of cells may be tested for their expression of specific antigens or their mRNA sequence intent. For example, a T-cell may be classified by virtue of its combination of expressed surface antigens. With specific reagents which interact with these antigens, a cell or a population of cells or a lysed cell may be exposed to a VLSIPS substrate. The biological sample may be classified or characterized by analyzing the pattern of specific interaction. This may be applicable to a cell or tissue type, to the messenger RNA population expressed by a cell to the genetic content of a cell, or to virtually any sample which can be classified and/or identified by its combination of specific molecular properties.
  • a species of organism may be characterized by its DNA sequences, e.g., a genetic fingerprint.
  • a fingerprinting method it may be determined that all members of that species are sufficiently similar in specific sequences that they can be easily identified as being within a particular group.
  • newly defined classes may be resolved by their similarity in fingerprint patterns.
  • a non-member of that group will fail to share those many identifying characteristics.
  • the technology allows testing of a very large number of specific interactions, it also provides the ability to more finely distinguish between closely related different cells or samples. This will have important applications in diagnosing viral, bacterial, and other pathological on nonpathological infections.
  • cell classification may be defined by any of a number of different properties.
  • a cell class may be defined by its DNA sequences contained therein. This allows species identification for parasitic or other infections.
  • the human cell is presumably genetically distinguishable from a monkey cell, but different human cells will share many genetic markers. At higher resolution, each individual human genome will exhibit unique sequences that can define it as a single individual.
  • a developmental stage of a cell type may be definable by its pattern of expression of messenger RNA. For example, in particular stages of cells, high levels of ribosomal RNA are found whereas relatively low levels of other types of messenger RNAs may be found. The high resolution distinguishability provided by this fingerprinting method allows the distinction between cells which have relatively minor differences in its expressed mRNA population. Where a pattern is shown to be characteristic of a stage, a stage may be defined by that particular pattern of messenger RNA expression.
  • the antigenic determinants found on a protein may very well define the cell class.
  • immunological T-cells are distinguishable from B-cells because, in part, the cell surface antigens on the cell types are distinguishable.
  • Different T-cell subclasses can be also distinguished from one another by whether they contain particular T-cell antigens.
  • the present invention provides the possibility for high resolution testing of many different interactions simultaneously, and the definition of new cell types will be possible.
  • the high resolution VLSIPSTM substrate may also be used as a very powerful diagnostic tool to test the combination of presence, of a plurality of different assays from a biological sample.
  • a cancerous condition may be indicated by a combination of various different properties found in the blood.
  • a cancerous condition may be indicated by a combination of expression of various soluble antigens found in the blood along with a high number of various cellular antigens found on lymphocytes and/or particular cell degradation products.
  • a substrate as provided herein a large number of different features can be simultaneously performed on a biological sample. In fact, the high resolution of the test will allow more complete characterization of parameters which define particular diseases.
  • the power of diagnostic tests may be limited by the extent of statistical correlation with a particular condition rather than with the number of antigens or interactions which are tested.
  • the present invention provides the means to generate this large universe of possible reagents and the ability to actually accumulate that correlative data.
  • a substrate as provided herein may be used for genetic screening. This would allow for simultaneous screening of thousands of genetic markers. As the density of the matrix is increased, many more molecules can be simultaneously tested. Genetic screening then becomes a simpler method as the present invention provides the ability to screen for thousands, tens of thousands, and hundreds of thousands, even millions of different possible genetic features. However, the number of high correlation genetic markers for conditions numbers only in the hundreds. Again, the possibility for screening a large number of sequences provides the opportunity for generating the data which can provide correlation between sequences and specific conditions or susceptibility. The present invention provides the means to generate extremely valuable correlations useful for the genetic detection of the causative mutation leading to medical conditions.
  • the present invention would be applicable to distinguishing two individuals having identical genetic compositions.
  • the antibody population within an individual is dependent both on genetic and historical factors. Each individual experiences a unique exposure to various infectious agents, and the combined antibody expression is partly determined thereby.
  • individuals may also be fingerprinted by their immunological content, either of actively expressed antibodies, or their immunological memory. Similar sorts of immunological and environmental histories may be useful for fingerprinting, perhaps in combination with other screening properties.
  • the present invention may be useful for screening allergic reactions or susceptibilities, and a simple IgE specificity test may be useful in determining a spectrum of allergies.
  • a cell sorter With the definition of new classes of cells, a cell sorter will be used to purify them. Moreover, new markers for defining that class of cells will be identified. For example, where the class is defined by its RNA content, cells may be screened by antisense probes which detect the presence or absence of specific sequences therein. Alternatively, cell lysates may provide information useful in correlating intracellular properties with extracellular markers which indicate functional differences. Using standard cell sorter technology with a fluorescence or labeled antisense probe which recognizes the internal presence of the specific sequences of interest, the cell sorter will be able to isolate a relatively homogeneous population of cells possessing the particular marker. Using successive probes the sorting process should be able to select for cells having a combination of a large number of different markers.
  • cells may be defined by the presence of other markers.
  • the markers may be carbohydrates, proteins, or other molecules.
  • a substrate having particular specific reagents, e.g., antibodies, attached to it should be able to identify cells having particular patterns of marker expression.
  • combinations of these made be utilized and a cell class may be defined by a combination of its expressed mRNA, its carbohydrate expression, its antigens, and other properties. This fingerprinting should be useful in determining the physiological state of a cell or population of cells.
  • these structural manifestations of function may be used to sort cells to generate a relatively homogeneous population of that class of cells.
  • Standard cell sorter technology may be applied to purify such a population, see, e.g., Dangl, J. and Herzenberg (1982) “Selection of hybridomas and hybridoma variants using the fluorescence activated cell sorter,” J. Immunological Methods 52:1-14; and Becton Dickinson, Fluorescence Activated Cell Sorter Division, San Jose, California, and Coulter Diagnostics, Hialeah, Florida.
  • an identification means arises from mosaicism problems in an organism.
  • a mosaic organism is one whose genetic content in different cells is significantly different.
  • Various clonal populations should have similar genetic fingerprints, though different clonal populations may have different genetic contents. See, for example, Suzuki et al. An Introduction to Genetic Analysis (4th Ed.), Freeman and Co., New York, which is hereby incorporated herein by reference.
  • this problem should be a relatively rare problem and could be more carefully evaluated with greater experience using the fingerprinting methods.
  • the invention will also find use in detecting changes, both genetic and antigenic, e.g., in a rapidly “evolving” protozoa infection, or similarly changing organism.
  • mapping parallels its use for fingerprinting and sequencing.
  • a polymer is a linear molecule
  • the mapping provides the ability to locate particular segments along the length of the polymer.
  • Branched polymers can be treated as a series of individual linear polymers.
  • the mapping provides the ability to locate, in a relative sense, the order of various subsequences. This may be achieved using at least two different approaches.
  • the first approach is to take the large sequence and fragment it at specific points.
  • the fragments are then ordered and attached to a solid substrate.
  • the clones resulting from a chromosome walking process may be individually attached to the substrate by methods, e.g., caged biotin techniques, indicated earlier. Segments of unknown map position will be exposed to the substrate and will hybridize to the segment which contains that particular sequence. This procedure allows the rapid determination of a number of different labeled segments, each mapping requiring only a single hybridization step once the substrate is generated. The substrate may be regenerated by removal of the interaction, and the next mapping segment applied.
  • a plurality of subsequences can be attached to a substrate.
  • Various short probes may be applied to determine which segments may contain particular overlaps.
  • the theoretical basis and a description of this mapping procedure is contained in, e.g., Evans et al. 1989 “Physical Mapping of Complex Genomes by Cosmid Multiplex Analysis,” Proc. Natl. Acad. Sci. USA 86:5030-5034, and other references cited above in the Section labeled “Overall Description.” Using this approach, the details of the mapping embodiment are very similar to those used in the fingerprinting embodiment.
  • the substrate may be generated in either of the methods generally applicable in the sequencing and fingerprinting embodiments.
  • the substrate may be made either synthetically, or by attaching otherwise purified probes or sequences to the matrix.
  • the probes or sequences may be derived either from synthetic or biological means.
  • the solid phase substrate synthetic methods may be utilized to generate a matrix with positionally defined sequences.
  • the importance of saturation of all possible subsequences of a preselected length is far less important than in the sequencing embodiment, but the length of the probes used may be desired to be much longer.
  • the processes for making a substrate which has longer oligonucleotide probes should not be significantly different from those described for the sequencing embodiments, but the optimization parameters may be modified to comply with the mapping needs.
  • the specificity of interaction between the targets and probe would typically be closer to those used for fingerprinting embodiments, where homology is more important than absolute distinguishability of high fidelity complementary hybridization.
  • the hybridization conditions will be such that merely homologous segments will interact and provide a positive signal.
  • it may be useful to measure the extent of homology by successive incubations at higher stringency conditions.
  • a plurality of different probes, each having various levels of homology may be used. In either way, the spectrum of homologies can be measured.
  • the specific interactions may also be compared in a fingerprint-like manner.
  • the specific reagents may have less specificity, e.g., monoclonal antibodies which recognize a broader spectrum of sequences may be utilized relative to a sequencing embodiment. Again, the specificity of interaction may be measured under various conditions of increasing stringency to determine the spectrum of matching across the specific probes selected, or a number of different stringency reagents may be included to indicate the binding affinity.
  • the detection methods used in the mapping procedure will be virtually identical to those used in the fingerprinting embodiment.
  • the detection methods will be selected in combination with the labeling methods.
  • mapping embodiment The analysis of the data in a mapping embodiment will typically be somewhat different from that in fingerprinting.
  • the fingerprinting embodiment will test for the presence or absence of specific or homologous segments.
  • the existence of an interaction is coupled with some indication of the location of the interaction.
  • the interaction is mapped in some manner to the physical polymer sequence.
  • Some means for determining the relative positions of different probes is performed. This may be achieved by synthesis of the substrate in pattern, or may result from analysis of sequences after they have been attached to the substrate.
  • the probes may be randomly positioned at various locations on the substrate.
  • the relative positions of the various reagents in the original polymer may be determined by using short fragments, e.g., individually, as target molecules which determine the proximity of different probes.
  • short fragments e.g., individually
  • target molecules which determine the proximity of different probes.
  • the substrate should be reusable in the manner described in the fingerprinting section.
  • the substrate is renewed by removal of the specific interactions and is washed and prepared for successive cycles of exposure to new target sequences.
  • mapping procedure may be used on other molecules than polynucleotides. Although hybridization is one type of specific interaction which is clearly useful for use in this mapping embodiment, antibody reagents may also be very useful. In the same way that polypeptide sequencing or other polymers may be sequenced by the reagents and techniques described in the sequencing section and fingerprinting section, the mapping embodiment may also be used similarly.
  • mapping in another form of mapping, as described above in the fingerprinting section, the developmental map of a cell or biological system may be measured using fingerprinting type technology. Thus, the mapping may be along a temporal dimension rather than along a polymer dimension.
  • the mapping or fingerprinting embodiments may also be used in determining the genetic rearrangements which may be genetically important, as in lymphocyte and B-cell development.
  • various rearrangements or chromosomal dislocations may be tested by either the fingerprinting or mapping methods. These techniques are similar in many respects and the fingerprinting and mapping embodiments may overlap in many respects.
  • oligonucleotides may be synthesized in automated fashion at specific locations on a matrix. In fact, these oligonucleotides may be used to direct other molecules to specific locations by linking specific oligonucleotides to other reagents which are in batch exposed to the matrix and hybridized in a complementary fashion to only those locations where the complementary oligonucleotide has been synthesized on the matrix.
  • the caged biotin method allows automated attachment, the speed of the caged biotin attachment process is relatively slow and requires a separate reaction for each reagent being attached.
  • the oligonucleotide method the specificity of position can be done in an automated and parallel fashion. As each reagent is produced, instead of directly attaching each reagent at each desired position, the reagent may be attached to a specific desired complementary oligonucleotide which will ultimately be specifically directed toward locations on the matrix having a complementary oligonucleotide attached thereat.
  • the technology allows screening for specificity of interaction with particular reagents.
  • the oligonucleotide sequence specificity of binding of a potential reagent may be tested by presenting to the reagent all of the possible subsequences available for binding.
  • secondary or higher order sequence specific features might not be easily screenable using this technology, it does provide a convenient, simple, quick, and thorough screen of interactions between a reagent and its target recognition sequences. See, e.g., Pfeifer et al. (1989) Science 246:810-812.
  • the interaction of a promoter protein with its target binding sequence may be tested for many different, or all, possible binding sequences.
  • the interaction of the promoter protein with each of the different potential binding sites may be analyzed.
  • the spectrum of strength of interactions with each different potential binding site may provide significant insight into the types of features which are important in determining specificity.
  • sequence specific interaction between reagents is the testing of binding of a double stranded nucleic acid structure with a single stranded oligonucleotide. Often, a triple stranded structure is produced which has significant aspects of sequence specificity. Testing of such interactions with either sequences comprising only natural nucleotides, or perhaps the testing of nucleotide analogs may be very important in screening for particularly useful diagnostic or therapeutic reagents. See, e.g., Haner and Dervan (1990) Biochemistry 29:9761-6765, and references therein.
  • the present invention provides a means to compare alleles or related sequences to locate and identify differences from the control sequence. This would be extremely useful in further analysis of genetic variability at a specific gene locus.
  • the present invention is also useful in defining specific stages in the temporal sequence of cells, e.g., development, and the resulting tissues within an organism.
  • the developmental stage of a cell, or population of cells can be dependent upon the expression of particular messenger RNAs or cellular antigens.
  • the screening procedures provided allow for high resolution definition of new classes of cells.
  • the temporal development of particular cells will be characterized by the presence or expression of various mRNAs. Means to simultaneously screen a plurality or very large number of different sequences are provided.
  • the combination of different markers made available dramatically increases the ability to distinguish fairly closely related cell types.
  • Other markers may be combined with markers and methods made available herein to define new classifications of biological samples, e.g., based upon new combinations of markers.
  • a plurality of antigens or specific binding proteins attached to the substrate may be used to define particular cell types.
  • subclasses of T-cells are defined, in part, by the combination of expressed cell surface antigens.
  • the present invention allows for the simultaneous screening of a large plurality of different antigens together.
  • higher resolution classification of different T-cell subclasses becomes possible and, with the definitions and functional differences which correlate with those antigenic or other parameters, the ability to purify those cell types becomes available. This is applicable not only to T-cells, but also to lymphocyte cells, or even to freely circulating cells. Many of the cells for which this would be most useful will be immobile cells found in particular tissues or organs.
  • Tumor cells will be diagnosed or detected using these fingerprinting techniques. Coupled with a temporal change in structure, developmental classes may also be selected and defined using these technologies.
  • the present invention also provides the ability not only to define new classes of cells based upon functional or structural differences, but it also provides the ability to select or purify populations of cells which share these particular properties. Standard cell sorting procedures using antibody markers may be used to detect extracellular features. Intracellular features would also be detectable by introducing the label reagents into the cell. In particular, antisense DNA or RNA molecules may be introduced into a cell to detect RNA sequences therein. See, e.g., Weintraub (1990) Scientific American 262:40-46.
  • the present invention also allows for the high resolution correlation of medical conditions with various different markers.
  • the presently available technology when applied to amniocentesis or other genetic screening methods, typically screens for tens of different markers at most.
  • the present invention allows simultaneous screening for tens, hundreds, thousands, tens of thousands, hundreds of thousands, and even millions of different genetic sequences.
  • applying the fingerprinting methods of the present invention to a sufficiently large population allows detailed statistical analysis to be made, thereby correlating particular medical conditions with particular markers, typically antigenic or genetic. Tumor specific antigens will be identified using the present invention.
  • the substrate is provided with a pattern of specific reagents which are positionally localized on the surface of the substrate. This matrix of positions is defined by the automated system which produces the substrate.
  • the instrument will typically be one similar to that described in Pirrung et al. (1992) U.S. Pat. No. 5,143,854, and Ser. No. 07/624,120, now abandoned.
  • the instrumentation described therein is directly applicable to the applications used here.
  • the apparatus comprises a substrate, typically a silicon containing substrate, on which positions on the surface may be defined by a coordinate system of positions. These positions can be individually addressed or detected by the VLSIPSTM Technology apparatus.
  • the VLSIPSTM Technology apparatus uses optical methods used in semiconductor fabrication applications. In this way, masks may be used to photo-activate positions for attachment or synthesis of specific sequences on the substrate. These manipulations may be automated by the types of apparatus described in Pirrung et al. (1992) U.S. Pat. No. 5,143,854 and Ser. No. 07/624,120, now abandoned.
  • Selectively removable protecting groups allow creation of well defined areas of substrate surface having differing reactivities.
  • the protecting groups are selectively removed from the surface by applying a specific activator, such as electromagnetic radiation of a specific wavelength and intensity. More preferably, the specific activator exposes selected areas of surface to remove the protecting groups in the exposed areas.
  • Protecting groups of the present invention are used in conjunction with solid phase oligomer syntheses, such as peptide syntheses using natural or unnatural amino acids, nucleotide syntheses using deoxyribonucleic and ribonucleic acids, oligosaccharide syntheses, and the like.
  • the protecting groups block a reactive end of the monomer to prevent self-polymerization. For instance, attachment of a protecting group to the amino terminus of an activated amino acid, such as the N-hydroxysuccinimide-activated ester of the amino acid prevents the amino terminus of one monomer from reacting with the activated ester portion of another during peptide synthesis.
  • the protecting group may be attached to the carboxyl group of an amino acid to prevent reaction at this site.
  • Most protecting groups can be attached to either the amino or the carboxyl group of an amino acid, and the nature of the chemical synthesis will dictate which reactive group will require a protecting group.
  • attachment of a protecting group to the 5′-hydroxyl group of a nucleoside during synthesis using for example, phosphate-triester coupling chemistry prevents the 5′-hydroxyl of one nucleoside from reacting with the 3′-activated phosphate-triester of another.
  • protecting groups are employed to protect a moiety on a molecule from reacting with another reagent.
  • Protecting groups of the present invention have the following characteristics: they prevent selected reagents from modifying the group to which they are attached; they are stable (that is, they remain attached) to the synthesis reaction conditions; they are removable under conditions that do not adversely affect the remaining structure; and once removed, do not react appreciably with the surface or surface-bound oligomer.
  • the selection of a suitable protecting group will depend, of course, on the chemical nature of the monomer unit and oligomer, as well as the specific reagents they are to protect against.
  • the protecting groups will be photoactivatable.
  • the properties and uses of photoreactive protecting compounds have been reviewed. See, McCray et al., Ann. Rev. of Biophys. and Biophys. Chem. (1989) 18:239-270, which is incorporated herein by reference.
  • the photosensitive protecting groups will be removable by radiation in the ultraviolet (UV) or visible portion of the electromagnetic spectrum. More preferably, the protecting groups will be removable by radiation in the near UV or visible portion of the spectrum. In some embodiments, however, activation may be performed by other methods such as localized heating, electron beam lithography, laser pumping, oxidation or reduction with microelectrodes, and the like.
  • Sulfonyl compounds are suitable reactive groups for electron beam lithography. Oxidative or reductive removal is accomplished by exposure of the protecting group to an electric current source, preferably using microelectrodes directed to the predefined regions of the surface which are desired for activation. A more detailed description of these protective groups is provided in Ser. No. 07/624,120, now abandoned, which is hereby incorporated herein by reference.
  • the density of reagents attached to a silicon substrate may be varied by standard procedures.
  • the surface area for attachment of reagents may be increased by modifying the silicon surface. For example, a matte surface may be machined or etched on the substrate to provide more sites for attachment of the particular reagents.
  • Another way to increase the density of reagent binding sites is to increase the derivitization density of the silicon. Standard procedures for achieving this are described, below.
  • One method to control the derivatization density is to highly derivatize the substrate with photochemical groups at high density.
  • the substrate is then photolyzed for various predetermined times, which photoactivate the groups at a measurable rate, and react them with a capping reagent.
  • the density of linker groups may be modulated by using a desired time and intensity of photoactivation.
  • the number of different sequences which may be provided may be limited by the density and the size of the substrate on which the matrix pattern is generated.
  • multiple substrates may be used to increase the number of sequences tested.
  • the number of sequences tested may be increased by using a plurality of different substrates. Because the VLSIPS apparatus is almost fully automated, increasing the number of substrates does not lead to a significant increase in the number of manipulations which must be performed by humans. This again leads to greater reproducibility and speed in the handling of these multiple substrates.
  • VLSIPSTM Technology generally allows a pattern or a matrix of reagents to be generated.
  • the procedure for making the pattern is performed by any of a number of different methods.
  • An apparatus and instrumentation useful for generating a high density VLSIPS substrate is described in detail in Pirrung et al. (1992) U.S. Pat. No. 5,143,854 and Ser. No. 07/624,120, now abandoned.
  • the binary masking technique allows for producing a plurality of sequences based on the selection of either of two possibilities at any particular location.
  • the binary decision may be the determination, on a particular synthetic cycle, whether or not to add any particular one of the possible subunits.
  • the binary masking strategy provides the ability to carry out spatially addressable parallel synthesis.
  • the synthetic methods in making a substrate are described in the parent application, Pirrung et al. (1992) U.S. Pat. No. 5,143,854.
  • the construction of the matrix pattern on the substrate will typically be generated by the use of photo-sensitive reagents.
  • photo-lithographic optical methods particular segments of the substrate can be irradiated with light to activate or deactivate blocking agents, e.g., to protect or deprotect particular chemical groups.
  • the substrates can have known polymers synthesized at positionally defined regions on the substrate. Methods for synthesizing various substrates are described in Pirrung et al. (1992) U.S. Pat. No.
  • nucleosides used in the synthesis of DNA by photolytic methods will typically be one of the two forms shown below:
  • B Adenine, Cytosine, Guanine, or Thymine
  • the photolabile group at the 5′ position is abbreviated UV (nitroveratryl) and in II, the group is abbreviated NVOC (nitroveratryl oxycarbonyl).
  • the bases adenine, cytosine, and guanine
  • NVOC nitrogenveratryl oxycarbonyl
  • the bases contain exocyclic NH 2 groups which must be protected during DNA synthesis.
  • Thymine contains no exocyclic NH 2 and therefore requires no protection.
  • the standard protecting groups for these amines are shown below:
  • R may be alkyl or aryl have been used.
  • FMOC 9-fluorenyl methoxycarbonyl
  • the advantage of the FMCC group is that it is removed under mild conditions (dilute organic bases) and can be used for all three bases.
  • the amide protecting groups require more harsh conditions to be removed (NH 3 /MeOH with heat)
  • Nucleosides used as 5′-OH probes useful in verifying correct VLSIPS synthetic function, include, for example, the following:
  • the method of attachment of the first nucleoside to the surface of the substrate depends on the functionality of the groups at the substrate surface. If the surface is amine functionalized, an amide bond is made (see example below).
  • Photolysis of the photolabile group NV or NVOC on the 5′ positions of the nucleosides is carried out at ⁇ 362 nm with an intensity of 14 mW/cm 2 for 10 minutes with the substrate side (side containing the photolabile group) immersed in dioxane. After the coupling of the next nucleoside is complete, the photolysis is repeated followed by another coupling until the desired oligomer is obtained.
  • the groups can be removed by mild base treatment 0.1N NaOH/MeOH or K 2 CO 3 /H 2 O/MeOH.
  • silyl ether Another group used most often is the silyl ether:
  • the nitroveratryl group could also be used to protect the 3′-position.
  • preformed polymers may be individually attached at particular sites on the substrate. This may be performed by individually attaching reagents one at a time to specific positions on the matrix, a process which may be automated. See, e.g., Ser. No. 07/435,316, now abandoned, and Barrett et al. (1993) U.S. Pat. No. 5,252,743.
  • Another way of generating a positionally defined matrix pattern on a substrate is to have individually specific reagents which interact with each specific position on the substrate. For example, oligonucleotides may be synthesized at defined locations on the substrate. Then the substrate would have on its surface a plurality of regions having homogeneous oligonucleotides attached at each position.
  • At least four different substrate preparation procedures are available for treating a substrate surface. They are the standard VLSIPSTM Technology method, polymeric substrates, DuraporeTM, and synthetic beads or fibers.
  • the treatment labeled “standard VLSIPSTM Technology” method is described in Ser. No. 07/624,120, now abandoned, and involves applying amino-propyltriethoxysilane to a glass surface.
  • the polymeric substrate approach involves either of two ways of generating a polymeric substrate.
  • the first uses a high concentration of aminopropyltriethoxysilane (2-20%) in an aqueous ethanol solution (95%). This allows the silane compound to polymerize both in solution and on the substrate surface, which provides a high density of amines on the surface of the glass. This density is contrasted with the standard VLSIPS method.
  • This polymeric method allows for the deposition on the substrate surface of a monolayer due to the anhydrous method used with the aforementioned silane.
  • the second polymeric method involves either the coating or covalent binding of an appropriate acrylic acid polymer onto the substrate surface.
  • an appropriate acrylic acid polymer e.g., in DNA synthesis, a monomer such as a hydroxypropylacrylate is used to generate a high density of hydroxyl groups on the substrate surface, allowing for the formation of phosphate bonds.
  • a monomer such as a hydroxypropylacrylate is used to generate a high density of hydroxyl groups on the substrate surface, allowing for the formation of phosphate bonds.
  • An example of such a compound is shown:
  • the method using a DuraporeTM membrane consists of a polyvinylidine difluoride coating with crosslinked polyhydroxylpropyl acrylate [PVDF-HPA]:
  • the fourth method utilizes synthetic beads or fibers. This would use another substrate, such as a teflon copolymer graft bead or fiber, which is covalently coated with an organic layer (hydrophilic) terminating in hydroxyl sites (commercially available from Molecular Biosystems, Inc.) This would offer the same advantage as the DuraporeTM membrane, allowing for immediate phosphate linkages, but would give additional contour by the 3-dimensional growth of oligomers.
  • a matrix pattern of new reagents may be targeted to each specific oligonucleotide position by attaching a complementary oligonucleotide to which the substrate bound form is complementary. For instance, a number of regions may have homogeneous oligonucleotides synthesized at various locations. Oligonucleotide sequences complementary to each of these can be individually generated and linked to a particular specific reagents. Often these specific reagents will be antibodies. As each of these is specific for finding its complementary oligonucleotide, each of the specific reagents will bind through the oligonucleotide to the appropriate matrix position.
  • a single step having a combination of different specific reagents being attached specifically to a particular oligonucleotide will thereby bind to its complement at the defined matrix position.
  • the oligonucleotides will typically then be covalently attached, using, e.g., an acridine dye, for photocrosslinking.
  • an acridine dye for photocrosslinking.
  • Psoralen is a commonly used acridine dye for photocrosslinking purposes, see, e.g., Song et al. (1979) Photochem. Photobiol. 29:1177-1197; Cimino et al. (1985) Ann. Rev. Biochem. 54:1151-1193; Parsons (1980) Photochem. Photobiol. 32:813-821; and Dattagupta et al.
  • antibody molecules may be used to specifically direct binding to defined positions on a substrate.
  • the VLSIPS technology may be used to generate specific epitopes at each position on the substrate.
  • Antibody molecules having specificity of interaction may be used to attach oligonucleotides, thereby avoiding the interference of internal polynucleotide sequences from binding to the substrate complementary oligonucleotides.
  • the specificity of interaction for positional targeting may be achieved by use of nucleotide analogues which do not interact with the natural nucleotides.
  • nucleotide analogues which do not interact with the natural nucleotides.
  • other synthetic nucleotides have been made which undergo base pairing, thereby providing the specificity of targeting, but the synthetic nucleotides also do not interact with the natural biological nucleotides.
  • VLSIPS synthetic processes would be useful in generating the VLSIPS substrate, and standard oligonucleotide synthesis could be applied, with minor modifications, to produce the complementary sequences which would be attached to other specific reagents.
  • an alternative method of attaching reagents in a positionally defined matrix pattern is to use a caged biotin system. See Barrett et al. (1993) U.S. Pat. No. 5,252,743, which is hereby incorporated herein by reference, for additional details on the chemistry and application of caged biotin embodiments.
  • the caged biotin has a photosensitive blocking moiety which prevents the combination of avidin to biotin. At positions where the photo-lithographic process has removed the blocking group, high affinity biotin sites are generated.
  • the surface immobilization may also take place by photo crosslinking of defined oligonucleotides linked to specific reagents. After hybridization of the complementary oligonucleotides, the oligonucleotides may be crosslinked by a reagent by psoralen or another similar type of acridine dye.
  • a reagent by psoralen or another similar type of acridine dye.
  • Other useful cross linking reagents are described in Dattagupta et al. (1985) U.S. Pat. No. 4,542,102, and (1987) U.S. Pat. No. 4,713,326.
  • colony or phage plaque transfer of biological polymers may be transferred directly onto a silicon substrate.
  • a colony plate may be transferred onto a substrate having a generic oligonucleotide sequence which hybridizes to another generic complementary sequence contained on all of the vectors into which inserts are cloned. This will specifically only bind those molecules which are actually contained in the vectors containing the desired complementary sequence. This immobilization allows for producing a matrix onto which a sequence specific reagent can bind, or for other purposes.
  • a plurality of different vectors each having a specific oligonucleotide attached to the vector may be specifically attached to particular regions on a matrix having a complementary oligonucleotide attached thereto.
  • the VLSIPSTM technology substrates may be used for screening for specific interactions with sequence specific targets or probes.
  • sequences having the entire repertoire of possible sequences of a defined length opens up the possibility of sequencing by hybridization.
  • This sequence may be de novo determination of an unknown sequence, particularly of nucleic acid, verification of a sequence determined by another method, or an investigation of changes in a previously sequenced gene, locating and identifying specific changes.
  • Maxam and Gilbert sequencing techniques are applied to sequences which have been determined by Sanger and Coulson.
  • Each of those sequencing technologies have problems with resolving particular types of sequences.
  • Sequencing by hybridization may serve as a third and independent method for verifying other sequencing techniques. See, e.g., (1988) Science 242:1245.
  • the ability to provide a large repertoire of particular sequences allows use of short subsequences and hybridization as a means to fingerprint a sample.
  • This may be used in a nucleic acid, as well as other polymer embodiments.
  • fingerprinting to a high degree of specificity of sequence matching may be used for identifying highly similar samples, e.g., those exhibiting high homology to the selected probes.
  • This may provide a means for determining classifications of particular sequences. This should allow determination of whether particular genomes of bacteria, phage, or even higher cells might be related to one another.
  • fingerprinting may be used to identify an individual source of biological sample. See, e.g., Lander, E. (1989) Nature, 339:501-505, and references therein.
  • a DNA fingerprint may be used to determine whether a genetic sample arose from another individual. This would be particularly useful in various sorts of forensic tests to determine, e.g., paternity or sources of blood samples.
  • Significant detail on the particulars of genetic fingerprinting for identification purposes are described in, e.g., Morris et al. (1989) “Biostatistical evolution of evidence from continuous allele frequency distribution DNA probes in reference to disputed paternity of identity,” J. Forensic Science 34:1311-1317; and Neufeld et al. (1990) Scientific American 262:46-53; each of which is hereby incorporated herein by reference.
  • a fingerprinting-like procedure may be used for classifying cell types by analyzing a pattern of specific nucleic acids present in the cell.
  • a series of antibodies may be used to identify cell markers, e.g., proteins, usually on the cell surface, but intracellular markers may also be used. Antigens which are extracellularly expressed are preferred so cell lysis is unnecessary in the screening, but intracellular markers may also be useful.
  • the markers will usually be proteins, but may be nucleic acids, lipids, metabolites, carbohydrates, or other cellular components. See, e.g., Winkelgren, I. (1990) Science News 136:234-237, which indicates extracellular DNA may be common, and suggesting that such might be characteristic of cell types, stage, or physiology.
  • stage of a cell, or group of cells may be tested or defined by isolating a sample of MRNA from the population and testing to see what sequences are present in messenger populations. Direct samples, or amplified samples, may be used. Where particular MRNA or other nucleic acid sequences may be characteristic of or shown to be characteristic of particular developmental stages, physiological states, or other conditions, this fingerprinting method may define them. Similar sorts of fingerprinting may be used for determining T-cell classes or perhaps even to generate classification schemes for such proteins as major histocompatibility complex antigens. Thus, the ability to make these substrates allows both the generation of reagents which will be used for defining subclasses or classes of cells or other biological materials, but also provides the mechanisms for selecting those cells which may be found in defined population groups.
  • the present invention In addition to cell classification defined by such a combination of properties, typically expression of extracellular antigens, the present invention also provides the means for isolating homogeneous population of cells. Once the antigenic determinants which define a cell class have been identified, these antigens may be used in a sequential selection process to isolate only those cells which exhibit the combination of defining structural properties.
  • the present invention may also be used for mapping sequences within a larger segment. This may be performed by at least two methods, particularly in reference to nucleic acids. Often, enormous segments of DNA are subcloned into a large plurality of subsequences. Ordering these subsequences may be important in determining the overlaps of sequences upon nucleotide determinations. Mapping may be performed by immobilizing particularly large segments onto a matrix using the VLSIPSTM Technology. Alternatively, sequences may be ordered by virtue of subsequences shared by overlapping segments. See, e.g., Craig et al. (1990) Nuc. Acids Res. 18:2653-2660; Michiels et al. (1987) CABIOS 3:203-210; and Olson et al. (1986) Proc. Natl. Acad. Sci. USA 83:7826-7830.
  • sequence specific reagents immobilized to the VLSIPSTM Technology substrate and another sequence specific reagent may be modified by the conditions of the interaction. Sequencing embodiments typically require high fidelity hybridization and the ability to discriminate perfect matching from imperfect matching. Fingerprinting and mapping embodiments may be performed using less stringent conditions, depending upon the circumstances.
  • the specificity of antibody/antigen interaction may depend upon such parameters as pH, salt concentration, ionic composition, solvent composition, detergent composition and concentration, and chaotropic agent concentration. See, e.g., Harlow and Lane (1988) Antibodies: A Laboratory Manual, Cold Spring Harbor Press, New York. By careful control of these parameters, the affinity of binding may be mapped across different sequences.
  • Parameters which are well known to affect specificity and kinetics of reaction include salt conditions, ionic composition of the solvent, hybridization temperature, length of oligonucleotide matching sequences, guanine and cytosine (GC) content, presence of hybridization accelerators, pH, specific bases found in the matching sequences, solvent conditions, and addition of organic solvents.
  • the salt conditions required for driving highly mismatched sequences to completion typically include a high salt concentration.
  • the typical salt used is sodium chloride (NaCl), however, other ionic salts may be utilized, e.g., KCl.
  • the salt concentration will often be less than about 3 molar, more often less than 2.5 molar, usually less than about 2 molar, and more usually less than about 1.5 molar.
  • the salt concentrations would typically be lower.
  • Ordinary high stringency conditions will utilize salt concentration of less than about 1 molar, more often less then about 750 millimolar, usually less than about 500 millimolar, and may be as low as about 250 or 150 millimolar.
  • the kinetics of hybridization and the stringency of hybridization both depend upon the temperature at which the hybridization is performed and the temperature at which the washing steps are performed. Temperatures at which steps for low stringency hybridization are desired would typically be lower temperatures, e.g., ordinarily at least about 15° C., more ordinarily at least about 20° C., usually at least about 25° C., and more usually at least about 30° C. For those applications requiring high stringency hybridization, or fidelity of hybridization and sequence matching, temperatures at which hybridization and washing steps are performed would typically be high. For example, temperatures in excess of about 35° C.
  • temperatures as high as 80° C., or even higher may be used.
  • the base composition of the specific oligonucleotides involved in hybridization affects the temperature of melting, and the stability of hybridization as discussed in the above references.
  • the bias of GC rich sequences to hybridize faster and retain stability at higher temperatures can be compensated for by the inclusion in the hybridization incubation or wash steps of various buffers.
  • Sample buffers which accomplish this result include the triethly-and trimethyl ammonium buffers. See, e.g., Wood et al. (1987) Proc. Natl. Acad. Sci. USA, 82:1585-1588, and Khrapko, K. et al. (1989) FEBS Letters 256:118-122.
  • the rate of hybridization can also be affected by the inclusion of particular hybridization accelerators.
  • hybridization accelerators include the volume exclusion agents characterized by dextran sulfate, or polyethylene glycol (PEG).
  • Dextran sulfate is typically included at a concentration of between 1% and 40% by weight. The actual concentration selected depends upon the application, but typically a faster hybridization is desired in which the concentration is optimized for the system in question. Dextran sulfate is often included at a concentration of between 0.5% and 2% by weight or dextran sulfate at a concentration between about 0.5% and 5%.
  • proteins which accelerate hybridization may be added, e.g., the recA protein found in E. coli or other homologous proteins.
  • the conditions of specific interaction would depend on the affinity of binding between the specific reagent and its target. Typically parameters which would be of particular importance would be pH, salt concentration anion and cation compositions, buffer concentration, organic solvent inclusion, detergent concentration, and inclusion of such reagents such as chaotropic agents.
  • the affinity of binding may be tested over a variety of conditions by multiple washes and repeat scans or by using reagents with differences in binding affinity to determine which reagents bind or do not bind under the selected binding and washing conditions.
  • the spectrum of binding affinities may provide an additional dimension of information which may be very useful in identification purposes and mapping.
  • the specific hybridization conditions will be selected to correspond to a discriminatory condition which provides a positive signal where desired but fails to show a positive signal at affinities where interaction is not desired. This may be determined by a number of titration steps or with a number of controls which will be run during the hybridization and/or washing steps to determine at what point the hybridization conditions have reached the stage of desired specificity.
  • Methods for detection depend upon the label selected. The criteria for selecting an appropriate label are discussed below, however, a fluorescent label is preferred because of its extreme sensitivity and simplicity. Standard labeling procedures are used to determine the positions where interactions between a sequence and a reagent take place. For example, if a target sequence is labeled and exposed to a matrix of different probes, only those locations where probes do interact with the target will exhibit any signal. Alternatively, other methods may be used to scan the matrix to determine where interaction takes place. Of course, the spectrum of interactions may be determined in a temporal manner by repeated scans of interactions which occur at each of a multiplicity of conditions. However, instead of testing each individual interaction separately, a multiplicity of sequence interactions may be simultaneously determined on a matrix.
  • the target polynucleotide may be labeled by any of a number of convenient detectable markers.
  • a fluorescent label is preferred because it provides a very strong signal with low background. It is also optically detectable at high resolution and sensitivity through a quick scanning procedure.
  • Other potential labeling moieties include, radioisotopes, chemiluminescent compounds, labeled binding proteins, heavy metal atoms, spectroscopic markers, magnetic labels, and linked enzymes.
  • Another method for labeling may bypass any label of the target sequence.
  • the target may be exposed to the probes, and a double strand hybrid is formed at those positions only. Addition of a double strand specific reagent will detect where hybridization takes place.
  • An intercalative dye such as ethidium bromide may be used as long as the probes themselves do not fold back on themselves to a significant extent forming hairpin loops. See, e.g., Sheldon et al. (1986) U.S. Pat. No. 4,582,789. However, the length of the hairpin loops in short oligonucleotide probes would typically be insufficient to form a stable duplex.
  • different targets may be simultaneously sequenced where each target has a different label. For instance, one target could have a green fluorescent label and a second target could have a red fluorescent label. The scanning step will distinguish sites of binding of the red label from those binding the green fluorescent label. Each sequence can be analyzed independently from one another.
  • Suitable chromogens will include molecules and compounds which absorb light in a distinctive range of wavelengths so that a color may be observed, or emit light when irradiated with radiation of a particular wave length or wave length range, e.g., fluorescers.
  • Biliproteins, e.g., phycoerythrin. may also serve as labels.
  • Suitable dyes are available, being primarily chosen to provide an intense color with minimal absorption by their surroundings.
  • Illustrative dye types include quinoline dyes, triarylmethane dyes, acridine dyes, alizarine dyes, phthaleins, insect dyes, azo dyes, anthraquinoid dyes, cyanine dyes, phenazathionium dyes, and phenazoxonium dyes.
  • fluorescers may be employed either by themselves or in conjunction with quencher molecules. Fluorescers of interest fall into a variety of categories having certain primary functionalities. These primary functionalities include 1- and 2-aminonaphthalene, p,p′-diaminostilbenes, pyrenes, quaternary phenanthridine salts, 9-aminoacridines, p,p′-diaminobenzophenone imines, anthracenes, oxacarbocyanine, merocyanine, 3-aminoequilenin, perylene, bis-benzoxazole, bis-p-oxazolyl benzene, 1,2-benzophenazin, retinol, bis-3-aminopyridinium salts, hellebrigenin, tetracycline, sterophenol, benzimidzaolylphenylamine, 2-oxo-3-chromen, indole, xanthen
  • Individual fluorescent compounds which have functionalities for linking or which can be modified to incorporate such functionalities include, e.g., dansyl chloride; fluoresceins such as 3,6-dihydroxy-9-phenylxanthhydrol; rhodamineisothiocyanate; N-phenyl 1-amino-8-sulfonatonaphthalene; N-phenyl 2-amino-6-sulfonatonaphthalene; 4-acetamido-4-isothiocyanato-stilbene-2,2′-disulfonic acid; pyrene-3-sulfonic acid; 2-toluidinonaphthalene-6-sulfonate; N-phenyl, N-methyl 2-aminoaphthalene-6-sulfonate; ethidium bromide; stebrine; auromine-0,2-(9′-anthroyl)palmitate; dansyl phosphatidylethanolamine; N,N′-dioct
  • fluorescers should absorb light above about 300 nm, preferably about 350 nm, and more preferably above about 400 nm, usually emitting at wavelengths greater than about 10 nm higher than the wavelength of the light absorbed. It should be noted that the absorption and emission characteristics of the bound dye may differ from the unbound dye. Therefore, when referring to the various wavelength ranges and characteristics of the dyes, it is intended to indicate the dyes as employed and not the dye which is unconjugated and characterized in an arbitrary solvent.
  • Fluorescers are generally preferred because by irradiating a fluorescer with light, one can obtain a plurality of emissions. Thus, a single label can provide for a plurality of measurable events.
  • Detectable signal may also be provided by chemiluminescent and bioluminescent sources.
  • Chemiluminescent sources include a compound which becomes electronically excited by a chemical reaction and may then emit light which serves as the detectible signal or donates energy to a fluorescent acceptor.
  • a diverse number of families of compounds have been found to provide chemiluminescence under a variety of conditions.
  • One family of compounds is 2,3-dihydro-1,-4-phthalazinedione.
  • the most popular compound is luminol, which is the 5-amino compound.
  • Other members of the family include the 5-amino-6,7,8-trimethoxy- and the dimethylamino[ca]benz analog.
  • Chemiluminescent analogs include para-dimethylamino and -methoxy substituents. Chemiluminescence may also be obtained with oxalates, usually oxalyl active esters, e.g., p-nitrophenyl and a peroxide, e.g., hydrogen peroxide, under basic conditions. Alternatively, luciferins may be used in conjunction with luciferase or lucigenins to provide bioluminescence.
  • Spin labels are provided by reporter molecules with an unpaired electron spin which can be detected by electron spin resonance (ESR) spectroscopy.
  • exemplary spin labels include organic free radicals, transitional metal complexes, particularly vanadium, copper, iron, and manganese, and the like.
  • exemplary spin labels include nitroxide free radicals.
  • the correlation of specific positional labeling is converted to the presence on the target of sequences for which the reagents have specificity of interaction.
  • the positional information is directly converted to a database indicating what sequence interactions have occurred.
  • the sequences which have interacted between the substrate matrix and the target molecule can be directly listed from the positional information.
  • the detection system used is described in Pirrung et al. (1992) U.S. Pat. No. 5,143,854; and Ser. No. 07/624,120, now abandoned.
  • the detection described therein is a fluorescence detector, the detector may be replaced by a spectroscopic or other detector.
  • the scanning system may make use of a moving detector relative to a fixed substrate, a fixed detector with a moving substrate, or a combination.
  • mirrors or other apparatus can be used to transfer the signal directly to the detector. See, e.g, Ser. No. 07/624,120, now abandoned, which is hereby incorporated herein by reference.
  • the detection method will typically also incorporate some signal processing to determine whether the signal at a particular matrix position is a true positive or may be a spurious signal. For example, a signal from a region which has actual positive signal may tend to spread over and provide a positive signal in an adjacent region which actually should not have one. This may occur, e.g., where the scanning system is not properly discriminating with sufficiently high resolution in its pixel density to separate the two regions. Thus, the signal over the spatial region may be evaluated pixel by pixel to determine the locations and the actual extent of positive signal. A true positive signal should, in theory, show a uniform signal at each pixel location. Thus, processing by plotting number of pixels with actual signal intensity should have a clearly uniform signal intensity. Regions where the signal intensities show a fairly wide dispersion, may be particularly suspect and the scanning system may be programmed to more carefully scan those positions.
  • the system can compare the possibilities for the next adjacent position and look at these in comparison with each other.
  • the system might be programmed to compare each of these possibilities and select that one which gives a strong positive. In this way, the system can also simultaneously provide some means of measuring the reliability of the determination by indicating what the average signal to background ratio actually is.
  • sequences which interact may be analyzed for their overlap regions and the original target sequence may be reconstructed from the collection of specific subsequences obtained therein.
  • Other sorts of analyses for different applications may also be performed, and because the scanning system directly interfaces with a computer the information need not be transferred manually. This provides for the ability to handle large amounts of data with very little human intervention. This, of course, provides significant advantages over manual manipulations. Increased throughput and reproducibility is thereby provided by the automation of a vast majority of steps in any of these applications.
  • Target sequence may not specifically correspond to any specific molecule, especially where the target sequence is broken and fragmented in the sequencing process, the sequence corresponds to a contiguous sequence of the subfragments.
  • the data analysis can be performed by a computer using an appropriate program. See, e.g., Drmanac, R. et al. (1989) Genomics 4:114-128; and a commercially available analysis program available from the Genetic Engineering Center, P.O. Box 794, 11000 Belgrade, Yugoslavia.
  • a sorting program to sort all of the subsequences using a defined hierarchy.
  • the hierarchy need not necessarily correspond to any physical hierarchy, but provides a means to determine, in order, which subfragments have actually been found in the target sequence. In this manner, overlaps can be checked and found directly rather than having to search throughout the entire set after each selection process.
  • the first 9 positions can be sorted.
  • a particular subsequence can be selected as in the examples, to determine where the process starts.
  • the sorting procedure provides the ability to immediately find the position of the subsequence which contains the first 9 positions and can compare whether there exists more than 1 subsequence during the first 9 positions.
  • the computer can easily generate all of the possible target sequences which contain given combination of subsequences. Typically there will be only one, but in various situations, there will be more.
  • FIG. 1 An exemplary flow chart for a sequencing program is provided in FIG. 1.
  • the program provides for automated scanning of the substrate to determine the positions of probe and target interaction. Simple processing of the intensity of the signal may be incorporated to filter out clearly spurious signals.
  • the positions with positive interaction are correlated with the sequence specificity of specific matrix positions, to generate the set of matching subsequences. This information is further correlated with other target sequence information, e.g., restriction fragment analysis.
  • the sequences are then aligned using overlap data, thereby leading to possible corresponding target sequences which will, optimally, correspond to a single target sequence.
  • a variety of computer systems may be used to run a sequencing program.
  • the program may be written to provide both the detecting and scanning steps together and will typically be dedicated to a particular scanning apparatus. However, the components and functional steps may be separated and the scanning system may provide an output, e.g., through tape or an electronic connection into a separate computer which separately runs the sequencing analysis program.
  • the computer may be any of a number of machines provided by standard computer manufacturers, e.g., IBM compatible machines, AppleTM machines, VAX machines, and others, which may often use a UNIXTM operating system. Of course, the hardware used to run the analysis program will typically determine what programming language would be used.
  • an exemplary embodiment is a polynucleotide sequence system.
  • the theoretical and mathematical manipulations necessary for data analysis of other linear molecules, such as polypeptides, carbohydrates, and various other polymers are conceptually similar.
  • Simple branching polymers will usually also be sequencable using similar technology.
  • additional recognition reagents be used to determine the nature and location of branches. This can easily be provided by use of appropriate specific reagents which would be generated by methods similar to those used to produce specific reagents for linear polymers.
  • a substrate is made with specific reagents that are relatively insensitive to the handling and processing steps involved in a single cycle of use, the substrate may often be reused.
  • the target molecules are usually stripped off of the solid phase specific recognition molecules.
  • the manipulations and conditions be selected as to be mild and to not affect the substrate. For example, if a substrate is acid labile, a neutral pH would be preferred in all handling steps. similar sensitivities would be carefully respected where recycling is desired.
  • the previously attached specific interaction would be disrupted and removed. This will typically involve exposing the substrate to conditions under which the interaction between probe and target is disrupted. Alternatively, it may be exposed to conditions where the target is destroyed. For example, where the probes are oligonucleotides and the target is a polynucleotide, a heating and low salt wash will often be sufficient to disrupt the interactions. Additional reagents may be added such as detergents, and organic or inorganic solvents which disrupt the interaction between the specific reagents and target. In an embodiment where the specific reagents are antibodies, the substrate may be exposed to a gentle detergent which will denature the specific binding between the antibody and its target. The conditions are selected to avoid severe disruption or destruction of the structure of the antibody and to maintain the specificity of the antibody binding site. Conditions with specific pH, detergent concentration, salt concentration, ionic concentration, and other parameters may be selected which disrupt the specific interactions.
  • the matrix will typically be maintained under conditions where the matrix itself and the linkages and specific reagents are preserved.
  • Various specific preservatives may be added which prevent degradation.
  • a preservative such as cyanide or azide may be added.
  • the chemical preservative should also be selected to preserve the chemical nature of the linkages and other components of the substrate.
  • a detergent may also be included.
  • a substrate comprising a large number of oligomers will be treated in a fashion which is known to maintain the quality and integrity of oligonucleotides.
  • These include storing the substrate in a carefully controlled environment under conditions of lower temperature, cation depletion (EDTA and EGTA), sterile conditions, and inert argon or nitrogen atmosphere.
  • nucleic acids may be easily sequenced by careful selection of the vectors and hosts used for amplifying and generating the specific target sequences.
  • vectors may be carefully selected having particular complementary sequences which are designed to attach to a genetic or specific oligomer on the substrate. This is also applicable to situations where it is desired to target particular sequences to specific locations on the matrix.
  • unnatural oligomers may be used to target natural probes to specific locations on the VLSIPS substrate.
  • particular probes may be generated for the mapping embodiment which are designed to have specific combinations of characteristics.
  • the construction of a mapping substrate may depend upon use of another automated apparatus which takes clones isolated from a chromosome walk and attaches them individually or in bulk to the VLSIPS substrate.
  • a variety of specific vectors having known and particular “targeting” sequences adjacent to the cloning sites may be individually used to clone a selected probe, and the isolated probe will then be targetable to a site on the VLSIPS substrate with a sequence complementary to the “target” sequence.
  • the selection of probes may be very important. Significant mathematical analysis may be applied to determine which specific sequences should be used as those probes. Of course, for fingerprinting use, these sequences would be most desired that show significant heterogeneity across the human population. Selection of the specific sequences which would most favorably be utilized will tend to be single copy sequences within the genome.
  • hybridization selections may be made for non-repetitive and single copy sequences. See, e.g., Britten and Kohne (1968) “Repeated Sequences in DNA,” Science 161:529-540.
  • various means for constructing an appropriate substrate may involve either mechanical or automated procedures.
  • the standard VLSIPS automated procedure involves synthesizing oligonucleotides or short polymers directly on the substrate.
  • Other circumstances may lend themselves to transfer a pattern from a petri plate onto a solid substrate.
  • sequencing may be performed either de novo or as a verification of another sequencing method.
  • the present hybridization technology provides the ability to sequence nucleic acids and polynucleotides de novo, or as a means to verify either the maxam and Gilbert chemical sequencing technique or Sanger and Coulson dideoxy- sequencing techniques.
  • the hybridization method is useful to verify sequencing determined by any other sequencing technique and to closely compare two similar sequences, e.g., to identify and locate sequence differences.
  • the present invention also provides means for sequencing other polymers. This includes polypeptides, carbohydrates, synthetic organic polymers, and other polymers. Again, the sequencing may be either verification or de novo.
  • sequencing can be very important in many different sorts of environments. For example, it will be useful in determining the genetic sequence of particular markers in various individuals.
  • polymers may be used as markers or for information containing molecules to encode information.
  • a short polynucleotide sequence may be included in large bulk production samples indicating the manufacturer, date, and location of manufacture of a product.
  • various drugs may be encoded with this information with a small number of molecules in a batch.
  • a pill may have somewhere from 10 to 100 to 1,000 or more very short and small molecules encoding this information.
  • this information may be decoded from a sample of the material using a polymerase chain reaction (PCR) or other amplification method.
  • PCR polymerase chain reaction
  • This encoding system may be used to provide the origin of large bulky samples without significantly affecting the properties of those samples.
  • chemical samples may also be encoded by this method thereby providing means for identifying the source and manufacturing details of lots.
  • the origin of bulk hydrocarbon samples may be encoded.
  • Production lots of organic compounds such as benzene or plastics may be encoded with a short molecule polymer.
  • Food stuffs may also be encoded using similar marking molecules. Even toxic waste samples can be encoded determining the source or origin. In this way, proper disposal can be traced or more easily enforced.
  • Similar sorts of encoding may be provided by fingerprinting-type analysis. Whether the resolution is absolute or less so, the concept of coding information on molecules such as nucleic acids, which can be amplified and later decoded, may be a very useful and important application.
  • This technology also provides the ability to include markers for origins of biological materials. For example, a patented animal line may be transformed with a particular unnatural sequence which can be traced back to its origin. With a selection of multiple markers, the likelihood could be negligible that a combination of markers would have independently arisen from a source other than the patented or specifically protected source. This technique may provide a means for tracing the actual origin of particular biological materials. Bacteria, plants, and animals will be subject to marking by such encoding sequences.
  • fingerprinting technology may also be used for data encryption. Moreover, fingerprinting allows for significant identification of particular individuals. Where the fingerprinting technology is standardized, and used for identification of large numbers of people, related equipment and peripheral processing will be developed to accompany the underlying technology. For example, specific equipment may be developed for automatically taking a biological sample and generating or amplifying the information molecules within the sample to be used in fingerprinting analysis. Moreover, the fingerprinting substrate may be mass produced using particular types of automatic equipment. Synthetic equipment may produce the entire matrix simultaneously by stepwise synthetic methods as provided by the VLSIPSTM technology. The attachment of specific probes onto a substrate may also be automated, e.g., making use of the caged biotin technology. See, e.g., Barrett et al.
  • peripheral processing may be important and may be dedicated to this specific application.
  • automated equipment for producing the substrates may be designed, or particular systems which take in a biological sample and output either a computer readout or an encoded instrument, e.g., a card or document which indicates the information and can provide that information to others.
  • An identification having a short magnetic strip with a few million bits may be used to provide individual identification and important medical information useful in a medical emergency.
  • data banks may be set up to correlate all of this information of fingerprinting with medical information. This may allow for the determination of correlations between various medical problems and specific DNA sequences. By collating large populations of medical records with genetic information, genetic propensities and genetic susceptibilities to particular medical conditions may be developed. Moreover, with standardization of substrates, the micro encoding data may be also standardized to reproduce the information from a centralized data bank or on an encoding device carried on an individual person. On the other hand, if the fingerprinting procedure is sufficiently quick and routine, every hospital may routinely perform a fingerprinting operation and from that determine many important medical parameters for an individual.
  • VLSIPS sequencing, fingerprinting, or mapping technology will be particularly appropriate.
  • agricultural livestock suppliers may be able to encode and determine whether their particular strains are being used by others.
  • the markers will indicate origin of genetic material. This is applicable to seed producers, livestock producers, and other suppliers of medical or agricultural biological materials.
  • polymers may be used to encode important information on source and batch and supplier. This is described in greater detail, e.g., “Applications of PCR to industrial problems,” (1990) in Chemical and Engineering News 68:145, which is hereby incorporated herein by reference.
  • the synthetic method can be applied to the storage of enormous amounts of information. Small substrates may encode enormous amounts of information, and its recovery will make use of the inherent replication capacity. For example, on regions of 10 ⁇ m ⁇ 10 ⁇ m, 1 cm 2 has 10 6 regions. In theory, the entire human genome could be attached in 1000 nucleotide segments on a 3 cm 2 surface. Genomes of endangered species may be stored on these substrates.
  • Fingerprinting may also be used for genetic tracing or for identifying individuals for forensic science purposes. See, e.g., Morris, J. et al. (1989) “Biostatistical Evaluation of Evidence From Continuous Allele Frequency Distribution DNA Probes in Reference to Disputed Paternity and Identity,” J. Forensic Science 34:1311-1317, and references provided therein; each of which is hereby incorporated herein by reference.
  • the high resolution fingerprinting allows the distinguishability to high resolution of particular samples.
  • new cell classifications may be defined based on combinations of a large number of properties. Similar applications will be found in distinguishing different species of animals or plants.
  • microbial identification may become dependent on characterization of the genetic content. Tumors or other cells exhibiting abnormal physiology will be detectable by use of the present invention. Also, knowing the genetic fingerprint of a microorganism may provide very useful information on how to treat an infection by such organism.
  • Modifications of the fingerprint embodiments may be used to diagnose the condition of the organism.
  • a blood sample is presently used for diagnosing any of a number of different physiological conditions.
  • a multi-dimensional fingerprinting method ade available by the present invention could become a routine means for diagnosing an enormous number of physiological features simultaneously. This may revolutionize the practice of medicine in providing information on an enormous number of parameters together at one time.
  • the genetic predisposition may also revolutionize the practice of medicine providing a physician with the ability to predict the likelihood of particular medical conditions arising at any particular moment. It also provides the ability to apply preventive medicine.
  • the present invention might also find application in use for screening new drugs and new reagents which may be very important in medical diagnosis or other applications. For example, a description of generating a population of monoclonal antibodies with defined specificities may be very useful for producing various drugs or diagnostic reagents.
  • kits with the reagents useful for performing sequencing, fingerprinting, and mapping procedures will have various compartments with the desired necessary reagents, e.g., substrate, labeling reagents for target samples, buffers, and other useful accompanying products.
  • the present invention also provides the means for mapping sequences within enormous stretches of sequence. For example, nucleotide sequences may be mapped within enormous chromosome size sequence maps. For example, it would be possible to map a chromosomal location within the chromosome which contains hundreds of millions of nucleotide base pairs.
  • the mapping and fingerprinting embodiments allow for testing of chromosomal translocations, one of the standard problems for which amniocentesis is performed.
  • the present invention provides a powerful tool and the means for performing sequencing, fingerprinting, and mapping functions on polymers. Although most easily and directly applicable to polynucleotides, polypeptides, carbohydrates, and other sorts of molecules can be advantageously utilized using the present technology.
  • the resulting irradiated mixtures were then analyzed by HPLC using a Varian MicroPak SP column (C 18 analytical) at a flow rate of 1 ml/min and a solvent system of 40% CH 3 CN and 60% water.
  • Thymidine has a retention time of 1.2 min and NVO-Thym-OH has a retention time of 2.1 min. It was seen that after 10 min of exposure the deprotection was complete.
  • the plate was then rinsed with acetonitrile, then oxidized with I 2 /H 2 O/THF/lutidine for 1 min, washed and dried.
  • the slide was then exposed to a 1:3 mixture of acetic anhydride:pyridine for 1 h, then washed and dried.
  • the substrate was then photolyzed in dioxane at 362 nm at 14 mW/cm 2 for 10 min using a 500 ⁇ m checkerboard mask, dried, and then treated with a mixture of the following:
  • the substrate which is, in a preferred embodiment, a glass substrate such as a microscope slide or cover slip.
  • a roughened surface will be useable but a plastic or other solid substrate is also appropriate.
  • the slide is soaked in an alkaline bath consisting of, e.g., 1 liter of 95% ethanol with 120 ml of water and 120 grams of sodium hydroxide for 12 hours. The slides are washed with a buffer and under running water, allowed to air dry, and rinsed with a solution of 95% ethanol.
  • the slides are then aminated with, e.g., aminopropyltriethoxysilane for the purpose of attaching amino groups to the glass surface on linker molecules, although other omega functionalized silanes could also be used for this purpose.
  • aminopropyltriethoxysilane is utilized, although solutions with concentrations from 10 ⁇ 2 % to 10% may be used, with about 10 ⁇ 3 % to 2% preferred.
  • a 0.1% mixture is prepared by adding to 100 ml of a 95% ethanol/5% water mixture, 100 microliters ( ⁇ l) of aminopropyltriethoxysilane.
  • the mixture is agitated at about ambient temperature on a rotary shaker for an appropriate amount of time, e.g., about 5 minutes. 500 ⁇ l of this mixture is then applied to the surface of one side of each cleaned slide. After 4 minutes or more, the slides are decanted of this solution and thoroughly rinsed three times or more by dipping in 100% ethanol.
  • the aminated surface of the slide is then exposed to about 500 ⁇ l of, for example, a 30 millimolar (mM) solution of NVOC-nucleotide-NHS (N-hydroxysuccinimide) in DMF for attachment of a NVOC-nucleotide to each of the amino groups.
  • mM millimolar
  • NVOC-nucleotide-NHS N-hydroxysuccinimide
  • any unreacted aminopropyl silane on the surface i.e., those amino groups which have not had the NVOC-nucleotide attached, are now capped with acetyl groups (to prevent further reaction) by exposure to a 1:3 mixture of acetic anhydride in pyridine for 1 hour.
  • Other materials which may perform this residual capping function include trifluoroacetic anhydride, formicacetic anhydride, or other reactive acylating agents.
  • the slides are washed again with DMF, methylene chloride, and ethanol.
  • FIG. 2 illustrates a possible synthesis of the eight trimers of the two-monomer set: cytosine and thymine (represented by C and T, respectively).
  • a glass slide bearing silane groups terminating in 6-nitroveratryloxycarboxamide (NVoC-NH) residues is prepared as a substrate.
  • Active esters penentafluorophenyl, OBt, etc.
  • cytosine and thymine protected at the 5′ hydroxyl group with NVOC are prepared as reagents. While not pertinent to this example, if side chain protecting groups are required for the monomer set, these must not be photoreactive at the wavelength of light used to protect the primary chain.
  • a cycle consists of:
  • the glass 20 is provided with regions 22 , 24 , 26 , 28 , 30 , 32 , 34 , and 36 .
  • Regions 30 , 32 , 34 , and 36 are masked, indicated by the hatched regions, as shown in FIG. 2B and the glass is irradiated by the bright regions 22 , 24 , 26 , and 28 , and exposed to a reagent containing a photosensitive blocked C (e.g., cytosine derivative), with the resulting structure shown in FIG. 2C.
  • a photosensitive blocked C e.g., cytosine derivative
  • regions 22 , 24 , 26 , and 28 are masked, as indicated by the hatched region, the glass is irradiated (as shown in FIG. 2D), as indicated by the bright regions, at 30 , 32 , 34 , and 36 , and exposed to a photosensitive blocked reagent containing T (e.g., thymine derivative), with the resulting structure shown in FIG. 2E.
  • T e.g., thymine derivative
  • the process proceeds, consecutively masking and exposing the sections as shown until the structure shown in FIG. 2M is obtained.
  • the glass is irradiated and the terminal groups are, optionally, capped by acetylation. As shown, all possible trimers of cytosine/thymine are obtained.
  • n the number of monomers in the basis set of monomers
  • the maximum number of lithographic steps needed will generally be n for each “layer” of monomers, i.e., the total number of masks (and, therefore, the number of lithographic steps) needed will be n ⁇ 1.
  • the size of the transparent mask regions will vary in accordance with the area of the substrate available for synthesis and the number of sequences to be formed. In general, the size of the synthesis areas will be:
  • A is the total area available for synthesis.
  • S is the number of sequences desired in the area.
  • the target oligonucleotide can be labeled using standard procedures referred to above.
  • a reagent which recognizes interaction e.g., ethidium bromide
  • fluorescence labeling techniques may be applied, see, e.g., Smith, et al. (1986) Nature, 321: 674-679; and Prober, et al. (1987) Science, 238:336-341. The techniques described therein will be followed with minimal modifications as appropriate for the label selected.
  • the described technique may be applied, with photosensitive blocked nucleotides corresponding to adenine, cytosine, guanine, and thymine, to make combinations of polynucleotides consisting of each of the four different nucleotides. All 16 possible dimers would be made using a minor modification of the described method.
  • the described technique for making dimers of A, C, G, and T may be further extended to make longer oligonucleotides.
  • the automated system described e.g., in Pirrung et al. (1992) U.S. Pat. No. 5,143,854, and Ser. No. 07/624,120, now abandoned, can be adapted to make all possible 10-mers composed of the 4 nucleotides A, C, G, and T.
  • the photosensitive, blocked nucleotide analogues have been described above, and would be readily adaptable to longer oligonucleotides.
  • the described hybridization conditions are directly applicable to the sequence specific recognition reagents attached to the substrate, produced as described immediately above.
  • the 10-mers have an inherent property of hybridizing to a complementary sequence.
  • the conditions of hybridization should be carefully selected, as described above. Careful control of the conditions, and titration of parameters should be performed to determine the optimum collective conditions.
  • Hybridization conditions are described in detail, e.g., in Hames and Higgins (1985) Nucleic Acid Hybridisation: A Practical Approach; and the considerations for selecting particular conditions are described, e.g., in Wetmur and Davidson, (1988) J. Mol. Biol. 31:349-370, and Wood et al. (1985) Proc. Natl. Acad. Sci. USA 82:1585-1588.
  • conditions are desired which can distinguish matching along the entire length of the probe from where there is one or more mismatched bases.
  • the length of incubation and conditions will be similar, in many respects, to the hybridization conditions used in Southern blot transfers.
  • the GC bias may be minimized by the introduction of appropriate concentrations of the alkylammonium buffers, as described above.
  • a fluorescently labeled target or set of targets are generated, as described in Prober, et al. (1987) Science 238:336-341, or Smith, et al. (1986) Nature 321:674-679.
  • the target or targets are of the same length as, or slightly longer, than the oligonucleotide probes attached to the substrate and they will have known sequences. Thus, only a few of the probes hybridize perfectly with the target, and which particular ones did would be known.
  • the substrate and probes are incubated under appropriate conditions for a sufficient period of time to allow hybridization to completion.
  • the time is measured to determine when the probe-target hybridizations have reached completion.
  • a salt buffer which minimizes GC bias is preferred, incorporating, e.g., buffer, such as tetramethyl ammonium or tetraethyl ammonium ion at between about 2.4 and 3.0 M. See Wood, et al. (1985) Proc. Nat'l Acad. Sci. USA 82:1585-1588.
  • This time is typically at least about 30 min, and may be as long as about 1-5 days. Typically very long matches will hybridize more quickly, very short matches will hybridize less quickly, depending upon relative target and probe concentrations.
  • the hybridization will be performed under conditions where the reagents are stable for that time duration.
  • the conditions for washing are titrated.
  • Three parameters initially titrated are time, temperature, and cation concentration of the wash step.
  • the matrix is scanned at various times to determine the conditions at which the distinguishability between true perfect hybrid and mismatched hybrid is optimized. These conditions will be preferred in the sequencing embodiments.
  • the detection of specific interactions may be performed by detecting the positions where the labeled target sequences are attached.
  • the label is a fluorescent label
  • the apparatus described e.g., in Pirrung et al. (1992) U.S. Pat. No. 5,143,854; and Ser. No. 07/624,120, now abandoned, may be advantageously applied.
  • the synthetic processes described above will result in a matrix pattern of specific sequences attached to the substrate, and a known pattern of interactions can be converted to corresponding sequences.
  • a separate reagent which differentially interacts with the probe and interacted probe/targets can indicate where interaction occurs or does not occur.
  • a single-strand specific reagent will indicate where no interaction has taken place, while a double-strand specific reagent will indicate where interaction has taken place.
  • An intercalating dye e.g., ethidium bromide, may be used to indicate the positions of specific interaction.
  • the aminated surface of the slide is exposed to about 500 ⁇ l of, e.g., a 30 millimolar (mM) solution of NVOC-GABA (gamma amino butyric acid) NES (N-hydroxysuccinimide) in DMF for attachment of a NVOC-GABA to each of the amino groups.
  • NVOC-GABA gamma amino butyric acid
  • NES N-hydroxysuccinimide
  • any unreacted aminopropyl silane on the surface i.e., those amino groups which have not had the NVOC-GABA attached, are now capped with acetyl groups (to prevent further reaction) by exposure to a 1:3 mixture of acetic anhydride in pyridine for 1 hour.
  • Other materials which may perform this residual capping function include trifluoroacetic anhydride, formicacetic anhydride, or other reactive acylating agents.
  • the slides are washed again with DMF, methylene chloride, and ethanol.
  • a functionalized DuraporeTM membrane was used as a substrate.
  • the DuraporeTM membrane was a polyvinylidine difluoride with aminopropyl groups.
  • the aminopropyl groups were protected with the DDZ group by reaction of the carbonyl chloride with the amino groups, a reaction readily known to those of skill in the art.
  • the surface bearing these groups was placed in a solution of THF and contacted with a mask bearing a checkerboard pattern of 1 mm opaque and transparent regions.
  • the mask was exposed to ultraviolet light having a wavelength down to at least about 280 nm for about 5 minutes at ambient temperature, although a wide range of exposure times and temperatures may be appropriate in various embodiments of the invention.
  • an exposure time of between about 1 and 5000 seconds may be used at process temperatures of between ⁇ 70 and +50° C.
  • exposure times of between about 1 and 500 seconds at about ambient pressure are used. In some preferred embodiments, pressure above ambient is used to prevent evaporation.
  • the surface of the membrane was then washed for about 1 hour with a fluorescent label which included an active ester bound to a chelate of a lanthanide. Wash times will vary over a wide range of values from about a few minutes to a few hours. These materials fluoresce in the red and the green visible region. After the reaction with the active ester in the fluorophore was complete, the locations in which the fluorophore was bound could be visualized by exposing them to ultraviolet light and observing the red and the green fluorescence. It was observed that the derivatized regions of the substrate closely corresponded to the original pattern of the mask.
  • the fluorescence intensity in all cases started off at a high value and then decreased exponentially.
  • the fall-off in intensity is due to photobleaching of the fluorescein molecules.
  • the traces of beads without fluorescein molecules are used for background subtraction.
  • the difference in the initial exponential decay between labeled and nonlabeled beads is integrated to give the total number of photon counts, and this number is related to the number of molecules per bead. Therefore, it is possible to deduce the number of photons per fluorescein molecule that can be detected. This calculation indicates the radiation of about 40 to 50 photons per fluorescein molecule are detected.
  • a slide with aminopropyl silane on its surface was immersed in a 1 mM solution of FITC in DMF for 1 hour at about ambient temperature. After reaction, the slide was washed twice with DMF and then washed with ethanol, water, and then ethanol again. It was then dried and stored in the dark until it was ready to be examined.
  • NVOC-GABA groups were attached as described above. The entire surface of one slide was exposed to light so as to expose a free amino group at the end of the gamma amino butyric acid. This slide, and a duplicate which was not exposed, were then exposed to fluorescein isothiocyanate (FITC).
  • FITC fluorescein isothiocyanate
  • FIG. 12A of Pirrung et al. (1992) U.S. Pat. No. 5,143,854 illustrates the slide which was not exposed to light, but which was exposed to FITC.
  • the units of the x axis are time and the units of the y axis are counts.
  • the trace contains a certain amount of background fluorescence.
  • the duplicate slide was exposed to 350 nm broadband illumination for about 1 minute (12 mW/cm 2 , ⁇ 350 nm illumination) , washed and reacted with FITC. A large increase in the level of fluorescence is observed, which indicates photolysis has exposed a number of amino groups on the surface of the slides for attachment of a fluorescent marker.
  • This slide was illuminated for approximately 5 minutes, with 12 mW of 350 nm broadband light and then reacted with the 1 mM FITC solution. It was put on the laser detection scanning stage and a graph was plotted as a two-dimensional representation of position color-coded for fluorescence intensity. The experiment was repeated a number of times through various masks.
  • the fluorescence patterns for a 100 ⁇ 100 ⁇ m mask, a 50 ⁇ m mask, a 20 ⁇ m mask, and a 10 ⁇ m mask indicate that the mask pattern is distinct down to at least about 10 ⁇ m squares using this lithographic technique.
  • Leu enkephalin was coupled to the surface and recognized by an antibody.
  • a slide was derivatized with 0.1% amino propyl-triethoxysilane and protected with NVOC.
  • a 500 ⁇ m checkerboard mask was used to expose the slide in a flow cell using backside contact printing.
  • the Leu enkephalin sequence H 2 N-tyrosine,glycine,glycine,phenylalanine,leucine-COOH, otherwise referred to herein as YGGFL
  • the peptide was added in DMF solution with the BOP/HOBT/DIEA coupling reagents and recirculated through the flow cell for 2 hours at room temperature.
  • a first antibody known as the Herz antibody
  • a second antibody goat anti-mouse fluorescein conjugate
  • a slide is derivatized with the aminopropyl group, protected in this case with t-BOC (t-butoxycarbonyl).
  • the slide was treated with TFA to remove the t-BOC protecting group.
  • E-aminocaproic acid which was t-BOC protected at its amino group, was then coupled onto the aminopropyl groups.
  • the aminocaproic acid serves as a spacer between the aminopropyl group and the peptide to be synthesized. The amino end of the spacer was deprotected and coupled to NVOC-leucine.
  • the entire slide was then illuminated with 12 mW of 325 nm broadband illumination.
  • the slide was then coupled with NVOC-phenylalanine and washed.
  • the resulting fluorescence scan showed dark areas containing the tetrapeptide GGFL, which is not recognized by the Herz antibody (and thus there is no binding of the goat anti-mouse antibody with fluorescein conjugate), and red areas in which YGGFL was present.
  • the YGGFL pentapeptide is recognized by the Herz antibody and, therefore, there is antibody in the lighted regions for the fluorescein-conjugated goat anti-mouse to recognize.
  • a fluorescence plot of the first slide, which contained only L- amino acids showed red areas (indicating strong binding, i.e., 149,000 counts or more) and black areas (indicating little or no binding of the Herz antibody, i.e., 20,000 counts or less).
  • the sequence YGGFL was clearly most strongly recognized.
  • the sequences YAGFL and YSGFL also exhibited strong recognition of the antibody. By contrast, most of the remaining sequences showed little or no binding.
  • the four duplicate portions of the slide were extremely consistent in the amount of binding shown therein.
  • a fluorescence plot of the D- amino acid slide indicated that strongest binding was exhibited by the YGGFL sequence. Significant binding was also detected to YaGFL, YsGFL, and YpGFL. The remaining sequences showed less binding with the antibody. Low binding efficiency of the sequence yGGFL was observed.
  • Table 6 lists the various sequences tested in order of relative fluorescence, which provides information regarding relative binding affinity. TABLE 6 Apparent Binding to Herz Ab L- a.a. Set D- a.a. Set YGGFL YGGFL YAGFL YaGFL YSGFL YsGFL LGGFL YpGFL FGGFL fGGFL YPGFL yGGFL LAGFL faGFL FAGFL wGGFL WGGFL yaGFL fpGFL waGFL
  • the methods provide for attaching to the surface a caged binding member which, in its caged form, has a relatively low affinity for other potentially binding species, such as receptors and specific binding substances.
  • a caged binding member which, in its caged form, has a relatively low affinity for other potentially binding species, such as receptors and specific binding substances.
  • the invention provides methods for forming predefined regions on a surface of a solid support, wherein the predefined regions are capable of immobilizing receptors.
  • the methods make use of caged binding members attached to the surface to enable selective activation of the predefined regions.
  • the caged binding members are liberated to act as binding members ultimately capable of binding receptors upon selective activation of the predefined regions.
  • the activated binding members are then used to immobilize specific molecules such as receptors on the predefined region of the surface.
  • the above procedure is repeated at the same or different sites on the surface so as to provide a surface prepared with a plurality of regions on the surface containing, for example, the same or different receptors.
  • receptors immobilized in this way have a differential affinity for one or more ligands, screenings and assays for the ligands can be conducted in the regions of the surface containing the receptors.
  • the alternative embodiment may make use of novel caged binding members attached to the substrate.
  • Caged (unactivated) members have a relatively low affinity for receptors of substances that specifically bind to uncaged binding members when compared with the corresponding affinities of activated binding members.
  • the binding members are protected from reaction until a suitable source of energy is applied to the regions of the surface desired to be activated.
  • a suitable energy source Upon application of a suitable energy source, the caging groups labilize, thereby presenting the activated binding member.
  • a typical energy source will be light.
  • the binding members on the surface may be attached to a receptor.
  • the receptor chosen may be a monoclonal antibody, a nucleic acid sequence, a drug receptor, etc.
  • the receptor will usually, though not always, be prepared so as to permit attaching it, directly or indirectly, to a binding member.
  • a specific binding substance having a strong binding affinity for the binding member and a strong affinity for the receptor or a conjugate of the receptor may be used to act as a bridge between binding members and receptors if desired.
  • the method uses a receptor prepared such that the receptor retains its activity toward a particular ligand.
  • the caged binding member attached to the solid substrate will be a photoactivatable biotin complex, i.e., a biotin molecule that has been chemically modified with photoactivatable protecting groups so that it has a significantly reduced binding affinity for avidin or avidin analogs than does natural biotin.
  • the protecting groups localized in a predefined region of the surface will be removed upon application of a suitable source of radiation to give binding members, that is biotin or a functionally analogous compound having substantially the same binding affinity for avidin or avidin analogs as does biotin.
  • avidin or an avidin analog is incubated with activated binding members on the surface until the avidin binds strongly to the binding members
  • the avidin so immobilized on predefined regions of the surface can then be incubated with a desired receptor or conjugate of a desired receptor.
  • the receptor will preferably be biotinylated, e.g., a biotinylated antibody, when avidin is immobilized on the predefined regions of the surface.
  • a preferred embodiment will present an avidin/biotinylated receptor complex, which has been previously prepared, to activated binding members on the surface.
  • Fingerprinting embodiments may be applied towards polynucleotide fingerprinting, polypeptide fingerprinting, cell and tissue classification, cell and tissue temporal development stage classification, diagnostic tests, forensic uses for individual identification, classification of organisms, and genetic screening of individuals. Mapping applications are also described below.
  • Polynucleotide fingerprinting may use reagents similar to those described above for probing a sequence for the presence of specific subsequences found therein.
  • the subsequences used for fingerprinting will be longer than the sequences used in oligonucleotide sequencing.
  • specific long segments may be used to determine the similarity of different samples of nucleic acids. They may also be used to fingerprint whether specific combinations of information are provided therein.
  • Particular probe sequences are selected and attached in a positional manner to a substrate. The means for attachment may be either using a caged biotin method described, e.g., in Barrett et al. (1993) U.S. Pat. No. 5,242,743, or by another method using targeting molecules.
  • a short polypeptide of specific sequence may be attached to an oligonucleotide and targeted to specific positions on a substrate having antibodies attached thereto, the antibodies exhibiting specificity for binding to those short peptide sequences.
  • an unnatural nucleotide or similar complementary binding molecule may be attached to the fingerprinting probe and the probe thereby directed towards complementary sequences on a VLSIPS substrate.
  • unnatural nucleotides would be preferred, e.g., unnatural optical isomers, which would not interfere with natural nucleotide interactions.
  • the substrate may be used in a manner quite similar to the sequencing embodiment to provide information as to whether the fingerprint probes are detecting the corresponding sequence in a target sequence. This will often provide information similar to a Southern blot hybridization.
  • a polypeptide fingerprint may be performed using antibodies which recognize specific antigens on the polypeptide.
  • monoclonal antibodies which recognize specific sequences or antigens on a polypeptide may be used to determine whether those epitopes are found on a particular protein. For example, particular patterns of epitopes would be found on various types of proteins. This will lead to the discovery that specific epitopes, or antigenic determinants, which are characteristic of, e.g., beta sheet segments, will be identified as will particular different types of domains in various protein types.
  • a screening method may be devised which can classify polypeptides, either native or denatured, into various new classes defined by the epitopes existing thereon.
  • a target peptide is exposed to the substrate.
  • the target may be either native or denatured, though the conditions used to denature the polypeptide may interfere with the specific interaction between the polypeptide and the recognition reagent.
  • This method is not dependent on the fact that the polypeptide is a single chain, thus protein complexes may also be fingerprinted using this methodology. Structures such as multi-subunit proteins, associations of proteins, ribosomes, nucleosomes, and other small cellular structures may also be fingerprinted and classified according to the presence of specific recognizable features thereon.
  • Peptide fingerprinting may be useful, for example, in correlating with particular physiological conditions or developmental stages of a cell or organism.
  • a biological sample may be fingerprinted to determine the presence in that sample of a plurality of different polypeptides which are each individually fingerprinted.
  • a polypeptide itself is not fingerprinted but a biological sample is fingerprinted searching for specific epitopes, e.g., polypeptide, carbohydrate, nucleic acid, or any of a number of other specific recognizable structural features.
  • the present invention can be used for cell classification using fingerprinting type technology as described above in the polypeptide fingerprint.
  • Classes of cells are typically defined by the presence of common functions which are usually reflected by structural features.
  • a plant cell is classified differently from an animal cell by a number of structural features.
  • the present invention provides improved means for distinguishing the different cell types.
  • T-cell classes are defined on the basis of expression of particular antigens characteristic of each class.
  • mouse T-cell differentiation markers include the LY antigens.
  • new populations and classes of cells may be defined.
  • different neural cell types may be defined on the basis of cell surface antigens.
  • tissue types will be defined on the basis of tissue specific antigens.
  • Developmental cell classes will be similarly defined. All of these screenings can make use of the VLSIPS substrates with specific recognition molecules attached thereto. The substrates are exposed to the cell types directly, assaying for attachment of cells to specific regions, or are exposed to products of a population of cells, e.g., a supernatant, or a cell lysate.
  • reagents which recognize those features may be developed and used in a fluorescence activated cell sorter as described, e.g., in Dangl, J. and Herzenberg (1982) J. Immunological Methods 52: 1-14; and Becton Dickinson, Fluorescence Activated Cell Sorters Division, San Jose, Calif. This will provide a homogeneous population of cells whose function has been defined by structure.
  • the present cell classification scheme may also be used to determine specific B-cell classes.
  • B-cells specific for producing IgM, IgG, IgD, IgE, and IgA may be defined by the internal expression of specific mRNA sequences encoding each type of immunoglobulin.
  • the classification scheme may depend on either extracellularly expressed markers which are correlated as being diagnostic of specific stages in development, or intracellular mRNA sequences which indicate particular functions.
  • the present fingerprinting invention also allows cell classification by expression of developmental antigens.
  • a lymphocyte stem cell expresses a particular combination of antigens. As the lymphocyte develops through a program developmental scheme, at various stages it expresses particular antigens which are diagnostic of particular stages in development.
  • the fingerprinting methodology allows for the definition of specific structural features which are diagnostic of developmental or functional features which will allow classification of cells into temporal developmental classes. Cells, products of those cells, or lysates of those cells will be assayed to determine the developmental stage of the source cells. In this manner, once a developmental stage is defined, specific synchronized populations of cells will be selected out of another population. These synchronized populations may be very important in determining the biological mechanisms of development.
  • the present invention also allows for fingerprinting of the mRNA population of a cell. In this fashion, the mRNA population, which should be a good determinant of developmental stage, will be correlated with other structural features of the cell. In this manner, cells at specific developmental stages will be characterized by the intracellular environment, as well as the extracellular environment.
  • the present invention also allows the combination of definitions based, in part, upon antigens and, in part, upon mRNA expression.
  • the two may be combined in a single incubation step.
  • a particular incubation condition may be found which is compatible with both hybridization recognition and non-hybridization recognition molecules.
  • an incubation condition may be selected which allows both specificity of antibody binding and specificity of nucleic acid hybridization. This allows simultaneous performance of both types of interactions on a single matrix.
  • a cell sorter may be used to sort specifically those cells having desired mRNA population patterns.
  • the present invention also provides the ability to perform diagnostic tests. Diagnostic tests typically are based upon a fingerprint type assay, which tests for the presence of specific diagnostic structural features. Thus, the present invention provides means for viral strain identification, bacterial strain identification, and other diagnostic tests using positionally defined specific reagents. The present invention also allows for determining a spectrum of allergies, diagnosing a biological sample for any or all of the above, and testing for many other conditions.
  • the present invention provides reagents and methodology for identifying viral strains.
  • the specific reagents may be either antibodies or recognition proteins which bind to specific viral epitopes preferably surface exposed, but may make use of internal epitopes, e.g., in a denatured viral sample.
  • the viral genome may be probed for specific sequences which are characteristic of particular viral strains. As above, a combination of the two may be performed simultaneously in a single interaction step, or in separate tests, e.g., for both genetic characteristics and epitope characteristics.
  • Similar techniques will be applicable to identifying a bacterial source. This may be useful in diagnosing bacterial infections, or in classifying sources of particular bacterial species.
  • the bacterial assay may be useful in determining the natural range of survivability of particular strains of bacteria across regions of the country or in different ecological niches.
  • the present invention provides means for diagnosis of other microbiological and other species, e.g., protozoal species and parasitic species in a biological sample, but also provides the means for assaying a combination of different infections.
  • a biological specimen may be assayed for the presence of any or all of these microbiological species.
  • typical samples will be blood, sputum, stool, urine, or other samples.
  • An immobilized set of antigens may be attached to a solid substrate and, instead of the standard skin reaction tests, a blood sample may be assayed on such a substrate to determine the presence of antibodies, e.g., IgE or other type antibodies, which may be diagnostic of an allergic or immunological susceptibility.
  • a standard radioallergosorbent test may be used to check a much larger population of antigens.
  • an allergy like test may be used to diagnose the immunological history of a particular individual. For example, by testing the circulating antibodies in a blood sample, which reflects the immunological history and memory of an individual, it may be determined what infections may not have been historically presented to the immune system. In this manner, it may be possible to specifically supplement an immune system for a short period of time with IgG fractions made up of specific types of gamma globulins. Thus, hepatitis gamma globulin injections may be better designed for a particular environment to which a person is expected to be exposed. This also provides the ability to identify genetically equivalent individuals who have immunologically different experiences.
  • a blood sample from an individual who has a particular combination of circulating antibodies will likely be different from the combination of circulating antibodies found in a genetically similar or identical individual. This could allow for the distinction between clones of particular animals, e.g., mice, rats, or other animals.
  • the present invention provides the ability to fingerprint and identify a genetic individual.
  • This individual may be a bacterial or lower microorganism, as described above in diagnostic tests, or of a plant or animal.
  • An individual may be identified genetically or immunologically, as described.
  • Genetic fingerprinting has been utilized in comparing different related species in Southern hybridization blots. Genetic fingerprinting has also been used in forensic studies, see, e.g., Morris et al. (1989) J. Forensic Science 34: 1311-1317, and references cited therein. As described above, an individual may be identified genetically by a sufficiently large number of probes. The likelihood that another individual would have an identical pattern over a sufficiently large number of probes may be statistically negligible. However, it is often quite important that a large number of probes be used where the statistical probability of matching is desired to be particularly low. In fact, the probes will optimally be selected for having high heterogeneity among the population.
  • the fingerprint method may make use of the pattern of homologies indicated by a series of more and more stringent washes. Then, each position has both a sequence specificity and a homology measurement, the combination of which greatly increases the number of dimensions and the statistical likelihood of a perfect pattern match with another genetic individual.
  • This same immunological screening may be used for other sorts of identifiable biological products.
  • an individual may be identified by her combination of expressed proteins. These proteins may reflect a physiological state of the individual, and would thus be useful in certain circumstances where diagnostic tests may be performed.
  • an individual may be identified, in part, by the presence of particular metabolic products.
  • a plant origin may be determined by virtue of having within its genome an unnatural sequence introduced to it by genetic breeders.
  • a marker nucleic acid sequence may be introduced as a means to determine whether a genetic strain of a plant or animal originated from another particular source.
  • the present invention provides for the ability to screen for genetic variations of individuals. For example, a number of genetic diseases are linked with specific alleles. See, e.g., Scriber, C. et al. (eds.) (1989) The Metabolic Bases of Inherited Disease, McGraw-Hill, New York. In one embodiment, cystic fibrosis has been correlated with a specific gene, see, Gregory et al. (1990) Nature 347: 382-386. A number of alleles are correlated with specific genetic deficiencies. See, e.g., McKusick, V.
  • amniocentesis is used to determine whether chromosome translocations have occurred.
  • the mapping procedure may provide the means for determining whether these translocations have occurred, and for detecting particular alleles of various markers.
  • the present invention allows for the positional location of specific clones useful for mapping.
  • caged biotin may be used for specifically positioning a probe to a location on a matrix pattern.
  • the specific probes may be positionally directed to specific locations on a substrate by targeting.
  • polypeptide specific recognition reagents may be attached to oligonucleotide sequences which can be complementarily targeted to specific locations on a VLSIPSTM Technology substrate. Hybridization conditions, as applied for oligonucleotide probes, will be used to target the reagents to locations on a substrate having complementary oligonucleotides synthesized thereon.
  • oligonucleotide probes may be attached to specific polypeptide targeting reagents such as an antigen or antibody. These reagents can be directed towards a complementary antigen or antibody already attached to a VLSIPS substrate.
  • an unnatural nucleotide which does not interfere with natural nucleotide complementary hybridization may be used to target oligonucleotides to particular positions on a substrate.
  • Unnatural optical isomers of natural nucleotides should be ideal candidates.
  • short probes may be used to determine the mapping of long targets or long targets may be used to map the position of shorter probes. See, e.g., Craig et al. 1990 Nuc. Acids Res. 18: 2653-2660.
  • Positionally defined clones may be transferred to a new substrate by either physical transfer or by synthetic means.
  • Synthetic means may involve either a production of the probe on the substrate using the VLSIPSTM Technology synthetic methods, or may involve the attachment of a targeting sequence made by VLSIPS synthetic methods which will target that positionally defined clone to a position on a new substrate. Both methods will provide a substrate having a number of positionally defined probes useful in mapping.
  • the present inventions provide greatly improved methods and apparatus for synthesis of polymers on substrates. It is to be understood that the above description is intended to be illustrative and not restrictive. Many embodiments will be apparent to those of skill in the art upon reviewing the above description. By way of example, the invention has been described primarily with reference to the use of photoremovable protective groups, but it will be readily recognized by those of skill in the art that sources of radiation other than light could also be used. For example, in some embodiments it may be desirable to use protective groups which are sensitive to electron beam irradiation, x-ray irradiation, in combination with electron beam lithograph, or x-ray lithography techniques. Alternatively, the group could be removed by exposure to an electric current. The scope of the invention should, therefore, be determined not with reference to the above description, but should instead be determined with reference to the appended claims, along with the full scope of equivalents to which such claims are entitled.

Abstract

The present invention provides methods and apparatus for sequencing, fingerprinting and mapping biological macromolecules, typically biological polymers. The methods make use of a plurality of sequence specific recognition reagents which can also be used for classification of biological samples, and to characterize their sources.

Description

    CROSS REFERENCE TO RELATED APPLICATIONS
  • This is a continuation of application Ser. No. 09/670,563, filed Sep. 27, 2000; which is a continuation of application Ser. No. 09/362,089, filed Jul. 28, 1999; which is a divisional of application Ser. No. 09/056,927, filed Apr. 8, 1998, pending; which is a continuation of application Ser. No. 08/670,118, filed Jun. 25, 1996, now U.S. Pat. No. 5,800,992; which is a divisional of application Ser. No. 08/168,904, filed Dec. 15, 1993; which is a continuation of application Ser. No. 07/624,114, filed Dec. 6, 1990, now abandoned; which is a continuation in-part of commonly assigned application Ser. No. 07/492,462, filed Mar. 7, 1990, now U.S. Pat. No. 5,143,854; and application Ser. No. 07/362,901, filed Jun. 7, 1989, now abandoned which are hereby incorporated by reference. [0001]
  • Additional commonly assigned application Ser. Nos. 07/624,120 and 07/626,730, both of which were filed on Dec. 6, 1990; application Ser. No. 07/435,316, filed Nov. 13, 1989, now abandoned; and U.S. Pat. No. 5,252,743 are also hereby incorporated herein by reference.[0002]
  • BACKGROUND OF THE INVENTION
  • The present invention relates to the sequencing, fingerprinting, and mapping of polymers, particularly biological polymers. The inventions may be applied, for example, in the sequencing, fingerprinting, or mapping of nucleic acids, polypeptides, oligosaccharides, and synthetic polymers. [0003]
  • The relationship between structure and function of macromolecules is of fundamental importance in the understanding of biological systems. These relationships are important to understanding, for example, the functions of enzymes, structural proteins, and signalling proteins, ways in which cells communicate with each other, as well as mechanisms of cellular control and metabolic feedback. [0004]
  • Genetic information is critical in continuation of life processes. Life is substantially informationally based and its genetic content controls the growth and reproduction of the organism and its complements. Polypeptides, which are critical features of all living systems, are encoded by the genetic material of the cell. In particular, the properties of enzymes, functional proteins, and structural proteins are determined by the sequence of amino acids which make them up. As structure and function are integrally related, many biological functions may be explained by elucidating the underlying structural features which provide those functions. For this reason, it has become very important to determine the genetic sequences of nucleotides which encode the enzymes, structural proteins, and other effectors of biological functions. In addition to segments of nucleotides which encode polypeptides, there are many nucleotide sequences which are involved in control and regulation of gene expression. [0005]
  • The human genome project is directed toward determining the complete sequence of the genome of the human organism. Although such a sequence would not correspond to the sequence of any specific individual, it would provide significant information as to the general organization and specific sequences contained within segments from particular individuals. It would also provide mapping information which is very useful for further detailed studies. However, the need for highly rapid, accurate, and inexpensive sequencing technology is nowhere more apparent than in a demanding sequencing project such as this. To complete the sequencing of a human genome would require the determination of approximately 3×10[0006] 9, or 3 billion base pairs.
  • The procedures typically used today for sequencing include the Sanger dideoxy method, see, e.g., Sanger et al. (1977) [0007] Proc. Natl. Acad. Sci. USA, 74:5463-5467, or the Maxam and Gilbert method, see, e.g., Maxam et al., (1980) Methods in Enzymology, 65:499-559. The Sanger method utilizes enzymatic elongation procedures with chain terminating nucleotides. The Maxam and Gilbert method uses chemical reactions exhibiting specificity of reaction to generate nucleotide specific cleavages. Both methods require a practitioner to perform a large number of complex manual manipulations. These manipulations usually require isolating homogeneous DNA fragments, elaborate and tedious preparing of samples, preparing a separating gel, applying samples to the gel, electrophoresing the samples into this gel, working up the finished gel, and analyzing the results of the procedure.
  • Thus, a less expensive, highly reliable, and labor efficient means for sequencing biological macromolecules is needed. A substantial reduction in cost and increase in speed of nucleotide sequencing would be very much welcomed. In particular, an automated system would improve the reproducibility and accuracy of procedures. The present invention satisfies these and other needs. [0008]
  • SUMMARY OF THE INVENTION
  • The present invention provides improved methods useful for de novo sequencing of an unknown polymer sequence, for verification of known sequences, for fingerprinting polymers, and for mapping homologous segments within a sequence. By reducing the number of manual manipulations required and automating most of the steps, the speed, accuracy, and reliability of these procedures are greatly enhanced. [0009]
  • The production of a substrate having a matrix of positionally defined regions with attached reagents exhibiting known recognition specificity can be used for the sequence analysis of a polymer. Although most directly applicable to sequencing, the present invention is also applicable to fingerprinting, mapping, and general screening of specific interactions. The VLSIPS™ Technology (Very Large Scale Immobilized Polymer Synthesis) substrates will be applied to evaluating other polymers, e.g., carbohydrates, polypeptides, hydrocarbon synthetic polymers, and the like. For these non-polynucleotides, the sequence specific reagents will usually be antibodies specific for a particular subunit sequence. [0010]
  • According to one aspect of the masking technique, the invention provides an ordered method for forming a plurality of polymer sequences by sequential addition of reagents comprising the step of serially protecting and deprotecting portions of the plurality of polymer sequences for addition of other portions of the polymer sequences using a binary synthesis strategy. [0011]
  • The present invention also provides a means to automate sequencing manipulations. The automation of the substrate production method and of the scan and analysis steps minimizes the need for human intervention. This simplifies the tasks and promotes reproducibility. [0012]
  • The present invention provides a composition comprising a plurality of positionally distinguishable sequence specific reagents attached to a solid substrate, which reagents are capable of specifically binding to a predetermined subunit sequence of a preselected multi-subunit length having at least three subunits, said reagents representing substantially all possible sequences of said preselected length. In some embodiments, the subunit sequence is a polynucleotide or a polypeptide, in others the preselected multi-subunit length is five subunits and the subunit sequence is a polynucleotide sequence. In other embodiments, the specific reagent is an oligonucleotide of at least about five nucleotides. Alternatively, the specific reagent is a monoclonal antibody. Usually the specific reagents are all attached to a single solid substrate, and the reagents comprise about 3000 different sequences. In other embodiments, the reagents represents at least about 25% of the possible subsequences of said preselected length. Usually, the reagents are localized in regions of the substrate having a density of at least 25 regions per square centimeter, and often the substrate has a surface area of less than about 4 square centimeters. [0013]
  • The present invention also provides methods for analyzing a sequence of a polynucleotide or a polypeptide, said method comprising the step of: [0014]
  • a) exposing said polynucleotide or polypeptide to a composition as described. [0015]
  • It also provides useful methods for identifying or comparing a target sequence with a reference, said method comprising the step of: [0016]
  • a) exposing said target sequence to a composition as described; [0017]
  • b) determining the pattern of positions of the reagents which specifically interact with the target sequence; and [0018]
  • c) comparing the pattern with the pattern exhibited by the reference when exposed to the composition. [0019]
  • The present invention also provides methods for sequencing a segment of a polynucleotide comprising the steps of: [0020]
  • a) combining: [0021]
  • i) a substrate comprising a plurality of chemically synthesized and positionally distinguishable oligonucleotides capable of recognizing defined oligonucleotide sequences; and [0022]
  • ii) a target polynucleotide; thereby forming high fidelity matched duplex structures of complementary subsequences of known sequence; and [0023]
  • b) determining which of said reagents have specifically interacted with subsequences in said target polynucleotide. [0024]
  • In one embodiment, the segment is substantially the entire length of said polynucleotide. [0025]
  • The invention also provides methods for sequencing a polymer, said method comprising the steps of: [0026]
  • a) preparing a plurality of reagents which each specifically bind to a subsequence of preselected length; [0027]
  • b) positionally attaching each of said reagents to one or more solid phase substrates, thereby producing substrates of positionally definable sequence specific probes; [0028]
  • c) combining said substrates with a target polymer whose sequence is to be determined; and [0029]
  • d) determining which of said reagents have specifically interacted with subsequences in said target polymer. [0030]
  • In one embodiment, the substrates are beads. Preferably, the plurality of reagents comprise substantially all possible subsequences of said preselected length found in said target. In another embodiment, the solid phase substrate is a single substrate having attached thereto reagents recognizing substantially all possible subsequences of preselected length found in said target. [0031]
  • In another embodiment, the method further comprises the step of analyzing a plurality of said recognized subsequences to assemble a sequence of said target polymer. In a bead embodiment, at least some of the plurality of substrates have one subsequence specific reagent attached thereto, and the substrates are coded to indicate the sequence specificity of said reagent. [0032]
  • The present invention also embraces a method of using a fluorescent nucleotide to detect interactions with oligonucleotide probes of known sequence, said method comprising: [0033]
  • a) attaching said nucleotide to a target unknown polynucleotide sequence, and [0034]
  • b) exposing said target polynucleotide sequence to a collection of positionally defined oligonucleotide probes of known sequences to determine the sequences of said probes which interact with said target. [0035]
  • In a further refinement, an additional step is included of: [0036]
  • a) collating said known sequences to determine the overlaps of said known sequences to determine the sequence of said target sequence. [0037]
  • A method of mapping a plurality of sequences relative to one another is also provided, the method comprising: [0038]
  • a) preparing a substrate having a plurality of positionally attached sequence specific probes; [0039]
  • b) exposing each of said sequences to said substrate, thereby determining the patterns of interaction between said sequence specific probes and said sequences; and [0040]
  • c) determining the relative locations of said sequence specific probe interactions on said sequences to determine the overlaps and order of said sequences. [0041]
  • In one refinement, the sequence specific probes are oligonucleotides, applicable to where the target sequences are nucleic acid sequences. [0042]
  • In the nucleic acid sequencing application, the steps of the sequencing process comprise: [0043]
  • a) producing a matrix substrate having known positionally defined regions of known sequence specific oligonucleotide probes; [0044]
  • b) hybridizing a target polynucleotide to the positions on the matrix so that each of the positions which contain oligonucleotide probes complementary to a sequence on the target hybridize to the target molecule; [0045]
  • c) detecting which positions have bound the target, thereby determining sequences which are found on the target; and [0046]
  • d) analyzing the known sequences contained in the target to determine sequence overlaps and assembling the sequence of the target therefrom. [0047]
  • The enablement of the sequencing process by hybridization is based in large part upon the ability to synthesize a large number (e.g., to virtually saturate) of the possible overlapping sequence segments and distinguishing those probes which hybridize with fidelity from those which have mismatched bases, and to analyze a highly complex pattern of hybridization results to determine the overlap regions. [0048]
  • The detecting of the positions which bind the target sequence would typically be through a fluorescent label on the target. Although a fluorescent label is probably most convenient, other sorts of labels, e.g., radioactive, enzyme linked, optically detectable, or spectroscopic labels may be used. Because the oligonucleotide probes are positionally defined, the location of the hybridized duplex will directly translate to the sequences which hybridize. Thus, analysis of the positions provides a collection of subsequences found within the target sequence. These subsequences are matched with respect to their overlaps so as to assemble an intact target sequence.[0049]
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 illustrates a flow chart for sequence, fingerprint, or mapping analysis. [0050]
  • FIG. 2A-M illustrates the process of a VLSIPS™ Technology trinucleotide synthesis.[0051]
  • DESCRIPTION OF THE PREFERRED EMBODIMENTS
  • I. Overall Description [0052]
  • A. general [0053]
  • B. VLSIPS substrates [0054]
  • C. binary masking [0055]
  • D. applications [0056]
  • E. detection methods and apparatus [0057]
  • F. data analysis [0058]
  • II. Theoretical Analysis [0059]
  • A. simple n-mer structure; theory [0060]
  • B. complications [0061]
  • C. non-polynucleotide embodiments [0062]
  • III. Polynucleotide Sequencing [0063]
  • A. preparation of substrate matrix [0064]
  • B. labeling target polynucleotide [0065]
  • C. hybridization conditions [0066]
  • D. detection; VLSIPS scanning [0067]
  • E. analysis [0068]
  • F. substrate reuse [0069]
  • G. non-polynucleotide aspects [0070]
  • IV. Fingerprinting [0071]
  • A. general [0072]
  • B. preparation of substrate matrix [0073]
  • C. labeling target nucleotides [0074]
  • D. hybridization conditions [0075]
  • E. detection; VLSIPS scanning [0076]
  • F. analysis [0077]
  • G. substrate reuse [0078]
  • H. non-polynucleotide aspects [0079]
  • V. Mapping [0080]
  • A. general [0081]
  • B. preparation of substrate matrix [0082]
  • C. labeling [0083]
  • D. hybridization/specific interaction [0084]
  • E. detection [0085]
  • F. analysis [0086]
  • G. substrate reuse [0087]
  • H. non-polynucleotide aspects [0088]
  • VI. Additional Screening [0089]
  • A. specific interactions [0090]
  • B. sequence comparisons [0091]
  • C. categorizations [0092]
  • D. statistical correlations [0093]
  • VII. Formation of Substrate [0094]
  • A. instrumentation [0095]
  • B. binary masking [0096]
  • C. synthetic methods [0097]
  • D. surface immobilization [0098]
  • VIII. Hybridization/Specific Interaction [0099]
  • A. general [0100]
  • B. important parameters [0101]
  • IX. Detection Methods [0102]
  • A. labeling techniques [0103]
  • B. scanning system [0104]
  • X. Data Analysis [0105]
  • A. general [0106]
  • B. hardware [0107]
  • C. software [0108]
  • XI. Substrate Reuse [0109]
  • A. removal of label [0110]
  • B. storage and preservation [0111]
  • C. processes to avoid degradation of oligomers [0112]
  • XII. Integrated Sequencing Strategy [0113]
  • A. initial mapping strategy [0114]
  • B. selection of smaller clones [0115]
  • C. actual sequencing procedures [0116]
  • XIII. Commercial Applications [0117]
  • A. sequencing [0118]
  • B. fingerprinting [0119]
  • C. mapping [0120]
  • I. Overall Description [0121]
  • A. General [0122]
  • The present invention relies in part on the ability to synthesize or attach specific recognition reagents at known locations on a substrate, typically a single substrate. In particular, the present invention provides the ability to prepare a substrate having a very high density matrix pattern of positionally defined specific recognition reagents. The reagents are capable of interacting with their specific targets while attached to the substrate, e.g., solid phase interactions, and by appropriate labeling of these targets, the sites of the interactions between the target and the specific reagents may be derived. Because the reagents are positionally defined, the sites of the interactions will define the specificity of each interaction. As a result, a map of the patterns of interactions with specific reagents on the substrate is convertible into information on the specific interactions taking place, e.g., the recognized features. Where the specific reagents recognize a large number of possible features, this system allows the determination of the combination of specific interactions which exist on the target molecule. Where the number of features is sufficiently large, the identical same combination, or pattern, of features is sufficiently unlikely that a particular target molecule may often be uniquely defined by its features. In the extreme, the features may actually be the subunit sequence of the target molecule, and a given target sequence may be uniquely defined by its combination of features. [0123]
  • In particular, the methodology is applicable to sequencing polynucleotides. The specific sequence recognition reagents will typically be oligonucleotide probes which hybridize with specificity to subsequences found on the target sequence. A sufficiently large number of those probes allows the fingerprinting of a target polynucleotide or the relative mapping of a collection of target polynucleotides, as described in greater detail below. [0124]
  • In the high resolution fingerprinting provided by a saturating collection of probes which include all possible subsequences of a given size, e.g., 10-mers, collating of all the subsequences and determination of specific overlaps will be derived and the entire sequence can usually be reconstructed. [0125]
  • Although a polynucleotide sequence analysis is a preferred embodiment, for which the specific reagents are most easily accessible, the invention is also applicable to analysis of other polymers, including polypeptides, carbohydrates, and synthetic polymers, including α-, β-, and ω-amino acids, polyurethanes, polyesters, polycarbonates, polyureas, polyamides, polyethyleneimines, polyarylene sulfides, polysiloxanes, polyimides, polyacetates, and mixed polymers. Various optical isomers, e.g., various D- and L- forms of the monomers, may be used. [0126]
  • Sequence analysis will take the form of complete sequence determination, to the level of the sequence of individual subunits along the entire length of the target sequence. Sequence analysis also takes the form of sequence homology, e.g., less than absolute subunit resolution, where “similarity” in the sequence will be detectable, or the form of selective sequences of homology interspersed at specific or irregular locations. [0127]
  • In either case, the sequence is determinable at selective resolution or at particular locations. Thus, the hybridization method will be useful as a means for identification, e.g., a “fingerprint”, much like a Southern hybridization method is used. It is also useful to map particular target sequences. [0128]
  • B. VLSIPS™ Technology [0129]
  • The invention is enabled by the development of technology to prepare substrates on which specific reagents may be either positionally attached or synthesized. In particular, the very large scale immobilized polymer synthesis (VLSIPS™) technology allows for the very high density production of an enormous diversity of reagents mapped out in a known matrix pattern on a substrate. These reagents specifically recognize subsequences in a target polymer and bind thereto, producing a map of positionally defined regions of interaction. These map positions are convertible into actual features recognized, and thus would be present in the target molecule of interest. [0130]
  • As indicated, the sequence specific recognition reagents will often be oligonucleotides which hybridize with fidelity and discrimination to the target sequence. For use with other polymers, monoclonal or polyclonal antibodies having high sequence specificity will often be used. [0131]
  • In the generic sense, the VLSIPS technology allows the production of a substrate with a high density matrix of positionally mapped regions with specific recognition reagents attached at each distinct region. By use of protective groups which can be positionally removed, or added, the regions can be activated or deactivated for addition of particular reagents or compounds. Details of the protection are described below and in related Pirrung et al. (1992) U.S. Pat. No. 5,143,854. In a preferred embodiment, photosensitive protecting agents will be used and the regions of activation or deactivation may be controlled by electro-optical and optical methods, similar to many of the processes used in semiconductor wafer and chip fabrication. [0132]
  • In the nucleic acid nucleotide sequencing application, a VLSIPS substrate is synthesized having positionally defined oligonucleotide probes. See Pirrung et al. (1992) U.S. Pat. No. 5,143,854; and U.S. Ser. No. 07/624,120, now abandoned. By use of masking technology and photosensitive synthetic subunits, the VLSIPS apparatus allows for the stepwise synthesis of polymers according to a positionally defined matrix pattern. Each oligonucleotide probe will be synthesized at known and defined positional locations on the substrate. This forms a matrix pattern of known relationship between position and specificity of interaction. The VLSIPS technology allows the production of a very large number of different oligonucleotide probes to be simultaneously and automatically synthesized including numbers in excess of about 10[0133] 2, 103, 104, 105, 106, or even more, and at densities of at least about 102, 103/cm2, 104/cm2, 105/cm2 and up to 106/cm2 or more. This application discloses methods for synthesizing polymers on a silicon or other suitably derivatized substrate, methods and chemistry for synthesizing specific types of biological polymers on those substrates, apparatus for scanning and detecting whether interaction has occurred at specific locations on the substrate, and various other technologies related to the use of a high density very large scale immobilized polymer substrate. In particular, sequencing, fingerprinting, and mapping applications are discussed herein in detail, though related technologies are described in simultaneously filed applications U.S. Ser. No. 07/624,120, now abandoned; and U.S. Ser. No. 07/517,659; Dower et al. (1995) U.S. Pat. No. 5,427,908, each of which is hereby incorporated herein by reference.
  • In other embodiments, antibody probes will be generated which specifically recognize particular subsequences found on a polymer. Antibodies would be generated which are specific for recognizing a three contiguous amino acid sequence, and monoclonal antibodies may be preferred. optimally, these antibodies would not recognize any sequences other than the specific three amino acid stretch desired and the binding affinity should be insensitive to flanking or remote sequences found on a target molecule. Likewise, antibodies specific for particular carbohydrate linkages or sequences will be generated. A similar approach could be used for preparing specific reagents which recognize other polymer subunit sequences. These reagents would typically be site specifically localized to a substrate matrix pattern where the regions are closely packed. [0134]
  • These reagents could be individually attached at specific sites on the substrate in a matrix by an automated procedure where the regions are positionally targeted by some other specific mechanism, e.g., one which would allow the entire collection of reagents to be attached to the substrate in a single reaction. Each reagent could be separately attached to a specific oligonucleotide sequence by an automated procedure. This would produce a collection of reagents where, e.g., each monoclonal antibody would have a unique oligonucleotide sequence attached to it. By virtue of a VLSIPS substrate which has different complementary oligonucleotides synthesized on it, each monoclonal antibody would specifically be bound only at that site on the substrate where the complementary oligonucleotide has been synthesized. A crosslinking step would fix the reagent to the substrate. See, e.g., Dattagupta et al. (1985) U.S. Pat. No. 4,542,102 and (1987) U.S. Pat. No. 4,713,326; and Chatterjee, M. et al. (1990) [0135] J. Am. Chem. Soc. 112:6397-6399, which are hereby incorporated herein by reference. This allows a high density positionally specific collection of specific recognition reagents, e.g., monoclonal antibodies, to be immobilized to a solid substrate using an automated system.
  • The regions which define particular reagents will usually be generated by selective protecting groups which may be activated or deactivated. Typically the protecting group will be bound to a monomer subunit or spatial region, and can be spatially affected by an activator, such as electromagnetic radiation. Examples of protective groups with utility herein include nitroveratryl oxycarbonyl (NVOC), nitrobenzyl oxycarbony (NBOC), dimethyl dimethoxy benzyloxy carbonyl, 5-bromo-7-nitroindolinyl, O-hydroxy-α-methyl cinnamoyl, and 2-oxymethylene anthraquinone. Examples of activators include ion beams, electric fields, magnetic fields, electron beams, x-ray, and other forms of electromagnetic radiation. [0136]
  • C. Binarv Masking [0137]
  • In fact, the means for producing a substrate useful for these techniques are explained in Pirrung et al. (1992) U.S. Pat. No. 5,143,854, which is hereby incorporated herein by reference. However, there are various particular ways to optimize the synthetic processes. Many of these methods are described in Ser. No. 07/624,120, now abandoned. [0138]
  • Briefly, the binary synthesis strategy refers to an ordered strategy for parallel synthesis of diverse polymer sequences by sequential addition of reagents which may be represented by a reactant matrix, and a switch matrix, the product of which is a product matrix. A reactant matrix is a 1×n matrix of the building blocks to be added. The switch matrix is all or a subset of the binary numbers from 1 to n arranged in columns. In preferred embodiments, a binary strategy is one in which at least two successive steps illuminate half of a region of interest on the substrate. In most preferred embodiments, binary synthesis refers to a synthesis strategy which also factors a previous addition step. For example, a strategy in which a switch matrix for a masking strategy halves regions that were previously illuminated, illuminating about half of the previously illuminated region and protecting the remaining half (while also protecting about half of previously protected regions and illuminating about half of previously protected regions). It will be recognized that binary rounds may be interspersed with non-binary rounds and that only a portion of a substrate may be subjected to a binary scheme, but will still be considered to be a binary masking scheme within the definition herein. A binary “masking” strategy is a binary synthesis which uses light to remove protective groups from materials for addition of other materials such as nucleotides or amino acids. [0139]
  • In particular, this procedure provides a simplified and highly efficient method for saturating all possible sequences of a defined length polymer. This masking strategy is also particularly useful in producing all possible oligonucleotide sequence probes of a given length. [0140]
  • D. Applications [0141]
  • The technology provided by the present invention has very broad applications. Although described specifically for polynucleotide sequences, similar sequencing, fingerprinting, mapping, and screening procedures can be applied to polypeptide, carbohydrate, or other polymers. In particular, the present invention may be used to completely sequence a given target sequence to subunit resolution. This may be for de novo sequencing, or may be used in conjunction with a second sequencing procedure to provide independent verification. See, e.g., (1988) [0142] Science 242:1245. For example, a large polynucleotide sequence defined by either the Maxam and Gilbert technique or by the Sanger technique may be verified by using the present invention.
  • In addition, by selection of appropriate probes, a polynucleotide sequence can be fingerprinted. Fingerprinting is a less detailed sequence analysis which usually involves the characterization of a sequence by a combination of defined features. Sequence fingerprinting is particularly useful because the repertoire of possible features which can be tested is virtually infinite. Moreover, the stringency of matching is also variable depending upon the application. A Southern Blot analysis may be characterized as a means of simple fingerprint analysis. [0143]
  • Fingerprinting analysis may be performed to the resolution of specific nucleotides, or may be used to determine homologies, most commonly for large segments. In particular, an array of oligonucleotide probes of virtually any workable size may be positionally localized on a matrix and used to probe a sequence for either absolute complementary matching, or homology to the desired level of stringency using selected hybridization conditions. [0144]
  • In addition, the present invention provides means for mapping analysis of a target sequence or sequences. Mapping will usually involve the sequential ordering of a plurality of various sequences, or may involve the localization of a particular sequence within a plurality of sequences. This may be achieved by immobilizing particular large segments onto the matrix and probing with a shorter sequence to determine which of the large sequences contain that smaller sequence. Alternatively, relatively shorter probes of known or random sequence may be immobilized to the matrix and a map of various different target sequences may be determined from overlaps. Principles of such an approach are described in some detail by Evans et al. (1989) “Physical Mapping of Complex Genomes by Cosmid Multiplex Analysis,” [0145] Proc. Natl. Acad. Sci. USA 86:5030-5034; Michiels et al. (1987) “Molecular Approaches to Genome Analysis: A Strategy for the Construction of Ordered Overlap Clone Libraries, ” CABIOS 3:203-210; Olsen et al. (1986) “Random-Clone Strategy for Genomic Restriction Mapping in Yeast,” Proc. Natl. Acad. Sci. USA 83:7826-7830; Craig, et al. (1990) “Ordering of Cosmid Clones Covering the Herpes Simplex Virus Type I (HSV-I) Genome: A Test Case for Fingerprinting by Hybridization, ” Nuc. Acids Res. 18:2653-2660; and Coulson, et al. (1986) “Toward a Physical Map of the Genome of the Nematode Caenorhabditis elegans, ” Proc. Natl. Acad. Sci. USA 83:7821-7825; each of which is hereby incorporated herein by reference.
  • Fingerprinting analysis also provides a means of identification. In addition to its value in apprehension of criminals from whom a biological sample, e.g., blood, has been collected, fingerprinting can ensure personal identification for other reasons. For example, it may be useful for identification of bodies in tragedies such as fire, flood, and vehicle crashes. In other cases the identification may be useful in identification of persons suffering from amnesia, or of missing persons. Other forensics applications include establishing the identity of a person, e.g., military identification “dog tags”, or may be used in identifying the source of particular biological samples. Fingerprinting technology is described, e.g., in Carrano, et al. (1989) “A High-Resolution, Fluorescence-Based, Semi-automated method for DNA Fingerprinting, ” [0146] Genomics 4: 129-136, which is hereby incorporated herein by reference. See, e.g., table I, for nucleic acid applications, and corresponding applications may be accomplished using polypeptides.
    TABLE I
    VLSIPS ™ TECHNOLOGY IN NUCLEIC ACIDS
    I. Construction of Chips
    II. Applications
    A. Sequencing
    1. Primary sequencing
    2. Secondary sequencing (sequence checking)
    3. Large scale mapping
    4. Fingerprinting
    B. Duplex/Triplex formation
    1. Antisense
    2. Sequence specific function modulation
    (e.g. promoter inhibition)
    C. Diagnosis
    1. Genetic markers
    2. Type markers
    a. Blood donors
    b. Tissue transplants
    D. Microbiology
    1. Clinical microbiology
    2. Food microbiology
    III. Instrumentation
    A. Chip machines
    B. Detection
    IV. Software Development
    A. Instrumentation software
    B. Data reduction software
    C. Sequence analysis software
  • The fingerprinting analysis may be used to perform various types of genetic screening. For example, a single substrate may be generated with a plurality of screening probes, allowing for the simultaneous genetic screening for a large number of genetic markers. Thus, prenatal or diagnostic screening can be simplified, economized, and made more generally accessible. [0147]
  • In addition to the sequencing, fingerprinting, and mapping applications, the present invention also provides means for determining specificity of interaction with particular sequences. Many of these applications were described in Ser. No. 07/362,901, now abandoned, Pirrung et al. (1992) U.S. Pat. No. 5,143,854; Ser. Nos. 07/435,316, and 07/612,671. [0148]
  • E. Detection Methods and Apparatus [0149]
  • An appropriate detection method applicable to the selected labeling method can be selected. Suitable labels include radionucleotides, enzymes, substrates, cofactors, inhibitors, magnetic particles, heavy metal atoms, and particularly fluorescers, chemiluminescers, and spectroscopic labels. Patents teaching the use of such labels include U.S. Pat. Nos. 3,817,837; 3,850,752; 3,939,350; 3,996,345; 4,277,437; 4,275,149; and 4,366,241. [0150]
  • With an appropriate label selected, the detection system best adapted for high resolution and high sensitivity detection may be selected. As indicated above, an optically detectable system, e.g., fluorescence or chemiluminescence would be preferred. other detection systems may be adapted to the purpose, e.g., electron microscopy, scanning electron microscopy (SEM), scanning tunneling electron microscopy (STEM), infrared microscopy, atomic force microscopy (AFM), electrical condutance, and image plate transfer. [0151]
  • With a detection method selected, an apparatus for scanning the substrate will be designed. Apparatus, as described in Ser. No. 07/362,901, now abandoned; or Pirrung et al. (1992) U.S. Pat. No. 5,143,854; or Ser. No. 07/624,120, now abandoned, are particularly appropriate. Design modifications may also be incorporated therein. [0152]
  • F. Data AnalVsis [0153]
  • Data is analyzed by processes similar to those described below in the section describing theoretical analysis. More efficient algorithms will be mathematically devised, and will usually be designed to be performed on a computer. Various computer programs which may more quickly or efficiently make measurement samples and distinguish signal from noise will also be devised. See, particularly, Ser. No. 07/624,120, now abandoned. [0154]
  • The initial data resulting from the detection system is an array of data indicative of fluorescent intensity versus location on the substrate. The data are typically taken over regions substantially smaller than the area in which synthesis of a given polymer has taken place. Merely by way of example, if polymers were synthesized in squares on the substrate having dimensions of 500 microns by 500 microns, the data may be taken over regions having dimensions of 5 microns by 5 microns. In most preferred embodiments, the regions over which florescence data are taken across the substrate are less than about ½ the area of the regions in which individual polymers are synthesized, preferably less than {fraction (1/10)} the area in which a single polymer is synthesized, and most preferably less than {fraction (1/100)} the area in which a single polymer is synthesized. Hence, within any area in which a given polymer has been synthesized, a large number of fluorescence data points are collected. [0155]
  • A plot of number of pixels versus intensity for a scan should bear a rough resemblance to a bell curve, but spurious data are observed, particularly at higher intensities. Since it is desirable to use an average of fluorescent intensity over a given synthesis region in determining relative binding affinity, these spurious data will tend to undesirably skew the data. [0156]
  • Accordingly, in one embodiment of the invention the data are corrected for removal of these spurious data points, and an average of the data points is thereafter utilized in determining relative binding efficiency. In general the data are fitted to a base curve and statistical measures are used to remove spurious data. [0157]
  • In an additional analytical tool, various degeneracy reducing analogues may be incorporated in the hybridization probes. Various aspects of this strategy are described, e.g., in Macevicz, S. (1990) PCT publication number WO 90/04652, which is hereby incorporated herein by reference. [0158]
  • II. Theoretical Analysis [0159]
  • The principle of the hybridization sequencing procedure is based, in part, upon the ability to determine overlaps of short segments. The VLSIPS technology provides the ability to generate reagents which will saturate the possible short subsequence recognition possibilities. The principle is most easily illustrated by using a binary sequence, such as a sequence of zeros and ones. Once having illustrated the application to a binary alphabet, the principle may easily be understood to encompass three letter, four letter, five or more letter, even 20 letter alphabets. A theoretical treatment of analysis of subsequence information to reconstruction of a target sequence is provided, e.e., in Lysov, Yu., et al. (1988) [0160] Doklady Akademi. Nauk. SSR 303:1508-1511; Khrapko K., et al. (1989) FEBS Letters 256:118-122; Pevzner, P. (1989) J. of Biomolecular Structure and Dynamics 7:63-69; and Drmanac, R. et al. (1989) Genomics 4:114-128; each of which is hereby incorporated herein by reference.
  • The reagents for recognizing the subsequences will usually be specific for recognizing a particular polymer subsequence anywhere within a target polymer. It is preferable that conditions may be devised which allow absolute discrimination between high fidelity matching and very low levels of mismatching. The reagent interaction will preferably exhibit no sensitivity to flanking sequences, to the subsequence position within the target, or to any other remote structure within the sequence. For polynucleotide sequencing, the specific reagents can be oligonucleotide probes; for polypeptides and carbohydrates, antibodies will be useful reagents. Antibody reagents should also be useful for other types of polymers. [0161]
  • A. Simple n-mer Structure: Theory [0162]
  • 1. Simple Two Letter Alphabet: Example [0163]
  • A simple example is presented below of how a sequence of ten digits comprising zeros and ones would be sequenceable using short segments of five digits. For example, consider the sample ten digit sequence: [0164]
  • 1010011100. [0165]
  • A VLSIPS™ Technology substrate could be constructed, as discussed elsewhere, which would have reagents attached in a defined matrix pattern which specifically recognize each of the possible five digit sequences of ones and zeros. The number of possible five digit subsequences is 2[0166] 5=32. The number of possible different sequences 10 digits long is 210=1,024. The five contiguous digit subsequences within a ten digit sequence number six, i.e., positioned at digits 1-5, 2-6, 3-7, 4-8, 5-9, and 6-10. It will be noted that the specific order of the digits in the sequence is important and that the order is directional, e.g., running left to right versus right to left. The first five digit sequence contained in the target sequence is 10100. The second is 01001, the third is 10011, the fourth is 00111, the fifth is 01110, and the sixth is 11100.
  • The VLSIPS™ substrate would have a matrix pattern of positionally attached reagents which recognize each of the different 5-mer subsequences. Those reagents which recognize each of the 6 contained 5-mers will bind the target, and a label allows the positional determination of where the sequence specific interaction has occurred. By correlation of the position in the matrix pattern, the corresponding bound subsequences can be determined. [0167]
  • In the above-mentioned sequence, six different 5-mer sequences would be determined to be present. They would be: [0168]
  • 10100 [0169]
  • 01001 [0170]
  • 10011 [0171]
  • 00111 [0172]
  • 01110 [0173]
  • 11100 [0174]
  • Any sequence which contains the first five digit sequence, 10100, already narrows the number of possible sequences (e.g., from 1024 possible sequences) which contain it to less than about 192 possible sequences. [0175]
  • This 192 is derived from the observation that with the subsequence 10100 at the far left of the sequence, in positions 1-5, there are only 32 possible sequences. Likewise, for that particular subsequence in positions 2-6, 3-7, 4-8, 5-9, and 6-10. So, to sum up all of the sequences that could contain 10100, there are 32 for each position and 6 positions for a total of about 192 possible sequences. However, some of these 10 digit sequences will have been counted twice. Thus, by virtue of containing the 10100 subsequence, the number of possible 10-mer sequences has been decreased from 1024 sequences to less than about 192 sequences. [0176]
  • In this example, not only do we know that the sequence contains 10100, but we also know that it contains the second five character sequence, 01001. By virtue of knowing that the sequence contains 10100, we can look specifically to determine whether the sequence contains a subsequence of five characters which contains the four leftmost digits plus a next digit to the left. For example, we would look for a sequence of X1010, but we find that there is none. Thus, we know that the 10100 must be at the left end of the 10-mer. We would also look to see whether the sequence contains the rightmost four digits plus a next digit to the right, e.g., 0100X. We find that the sequence also contains the sequence 01001, and that X is a 1. Thus, we know at least that our target sequence has an overlap of 0100 and has the left terminal sequence 101001. [0177]
  • Applying the same procedure to the second 5-mer, we also know that the sequence must include a sequence of five digits having the sequence 1001Y where Y must be either 0 or 1. We look through the fragments and we see that we have a 10011 sequence within our target, thus Y is also 1. Thus, we would know that our sequence has a sequence of the first seven being 1010011. [0178]
  • Moving to the next 5-mer, we know that there must be a sequence of 0011Z, where Z must be either 0 or 1. We look at the fragments produced above and see that the target sequence contains a 00111 subsequence and Z is 1. Thus, we know the sequence must start with 10100111. [0179]
  • The next 5-mer must be of the sequence 0111W where W must be 0 or 1. Again, looking up at the fragments produced, we see that the target sequence contains a 01110 subsequence, and W is a 0. Thus, our sequence to this point is 101001110. We know that the last 5-mer must be either 11100 or 11101. Looking above, we see that it is 11100 and that must be the last of our sequence. Thus, we have determined that our sequence must have been 1010011100. [0180]
  • However, it will be recognized from the example above with the sequences provided therein, that the sequence analysis can start with any known positive probe subsequence. The determination may be performed by moving linearly along the sequence checking the known sequence with a limited number of next positions. Given this possibility, the sequence may be determined, besides by scanning all possible oligonucleotide probe positions, by specifically looking only where the next possible positions would be. This may increase the complexity of the scanning but may provide a longer time span dedicated towards scanning and detecting specific positions of interest relative to other sequence possibilities. Thus, the scanning apparatus could be set up to work its way along a sequence from a given contained oligonucleotide to only look at those positions on the substrate which are expected to have a positive signal. [0181]
  • It is seen that given a sequence, it can be de-constructed into n-mers to produce a set of internal contiguous subsequences. From any given target sequence, we would be able to determine what fragments would result. The hybridization sequence method depends, in part, upon being able to work in the reverse, from a set of fragments of known sequences to the full sequence. In simple cases, one is able to start at a single position and work in either or both directions towards the ends of the sequence as illustrated in the example. [0182]
  • The number of possible sequences of a given length increases very quickly with the length of that sequence. Thus, a 10-mer of zeros and ones has 1024 possibilities, a 12-mer has 4096. A 20-mer has over a million possibilities, and a 30-mer has over a billion. However, a given 30-mer has, at most, 26 different internal 5-mer sequences. Thus, a 30 character target sequence having over a million possible sequences can be substantially defined by only 26 different 5-mers. It will be recognized that the probe oligonucleotides will preferably, but need not necessarily, be of identical length, and that the probe sequences need not necessarily be contiguous in that the overlapping subsequences need not differ by only a single subunit. Moreover, each position of the matrix pattern need not be homogeneous, but may actually contain a plurality of probes of known sequence. In addition, although all of the possible subsequence specifications would be preferred, a less than full set of sequences specifications could be used. In particular, although a substantial fraction will preferably be at least about 70%, it may be less than that. About 20% would be preferred, more preferably at least about 30% would be desired. Higher percentages would be especially preferred. [0183]
  • 2. Example of Four Letter Alphabet [0184]
  • A four letter alphabet may be conceptualized in at least two different ways from the two letter alphabet. One way is to consider the four possible values at each position and to analogize in a similar fashion to the binary example each of the overlaps. A second way is to group the binary digits into groups. [0185]
  • Using the first means, the overlap comparisons are performed with a four letter alphabet rather than a two letter alphabet. Then, in contrast to the binary system with 10 positions where 2[0186] 10=1024 possible sequences, in a 4-character alphabet with 10 positions, there will actually be 410=1,048,576 possible sequences. Thus, the complexity of a four character sequence has a much larger number of possible sequences compared to a two character sequence. Note, however, that there are still only 6 different internal 5-mers. For simplicity, we shall examine a 5 character string with 3 character subsequences. Instead of only 1 and 0, the characters may be designated, e.g., A, C, G, and T. Let us take the sequence GGCTA. The 3-mer subsequences are:
  • GGC [0187]
  • GCT [0188]
  • CTA [0189]
  • Given these subsequences, there is one sequence, or at most only a few sequences which would produce that combination of subsequences, i.e., GGCTA. [0190]
  • Alternatively, with a four character universe, the binary system can be looked at in pairs of digits. The pairs would be 00, 01, 10, and 11. In this manner, the earlier used sequence 1010011100 is looked at as 10, 10, 01, 11, 00. Then the first character of two digits is selected from the possible universe of the four representations 00, 01, 10, and 11. Then a probe would be in an even number of digits, e.g., not five digits, but, three pairs of digits or six digits. A similar comparison is performed and the possible overlaps determined. The 3-pair subsequences are: [0191]
  • 10, 10, 01 [0192]
  • 10, 01, 11 [0193]
  • 01, 11, 00 [0194]
  • and the overlap reconstruction produces 10, 10, 01, 11, 00. [0195]
  • The latter of the two conceptual views of the 4 letter alphabet provides a representation which is similar to what would be provided in a digital computer. The applicability to a four nucleotide alphabet is easily seen by assigning, e.g., 00 to A, 01 to C, 10 to G, and 11 to T. And, in fact, if such a correspondence is used, both examples for the 4 character sequences can be seen to represent the same target sequence. The applicability of the hybridization method and its analysis for determining the ultimate sequence is easily seen if A is the representation of adenine, C is the representation of cytosine, G is the representation of guanine, and T is the representation of thymine or uracil. [0196]
  • 3. Generalization to m-letter Alphabet [0197]
  • This reconstruction process may be applied to polymers of virtually any number of possible characters in the alphabet, and for virtually any length sequence to be sequenced, though limitations, as discussed below, will limit its efficiency at various extremes of length. It will be recognized that the theory can be applied to a large diversity of systems where sequence is important. [0198]
  • For example, the method could be applied to sequencing of a polypeptide. A polypeptide can have any of twenty natural amino acid possibilities at each position. A twenty letter alphabet is amenable to sequencing by this method so long as reagents exist for recognizing shorter subsequences therein. A preferred reagent for achieving that goal would be a set of monoclonal antibodies each of which recognizes a specific three contiguous amino acid subsequence. A complete set of antibodies which recognize all possible subsequences of a given length, e.g., 3 amino acids, and preferably with a uniform affinity, would be 20[0199] 3=8000 reagents.
  • It will also be recognized that each target sequence which is recognized by the specific reagents need not have homogeneous termini. Thus, fragments of the entire target sequence will also be useful for hybridizing appropriate subsequences. It is, however, preferable that there not be a significant amount of labeled homogeneous contaminating extraneous sequences. This constraint does usually require the purification of the target molecule to be sequenced, but a specific label technique would dispense with a purification requirement if the unlabeled extraneous sequences do not interfere with the labeled sequences. [0200]
  • In addition, conformational effects of target polypeptide folding may, in certain embodiments, be negligible if the polypeptide is fragmented into sufficiently small peptides, or if the interaction is performed under conditions where conformation, but not specific interaction, is disrupted. [0201]
  • B. Complications [0202]
  • Two obvious complications exist with the method of sequence analysis by hybridization. The first results from a probe of inappropriate length while the second relates to internally repeated sequences. [0203]
  • The first obvious complication is a problem which arises from an inappropriate length of recognition sequence, which causes problems with the specificity of recognition. For example, if the recognized sequence is too short, every sequence which is utilized will be recognized by every probe sequence. This occurs, e.g., in a binary system where the probes are each of sequences which occur relatively frequently, e.g., a two character probe for the binary system. Each possible two character probe would be expected to appear ¼ of the time in every single two character position. Thus, the above sequence example would be recognized by each of the 00, 10, 01, and 11. Thus, the sequence information is virtually lost because the resolution is too low and each recognition reagent specifically binds at multiple sites on the target sequence. [0204]
  • The number of different probes which bind to a target depends on the relationship between the probe length and the target length. At the extreme of short probe length, the just mentioned problem exists of excessive redundancy and lack of resolution. The lack of stability in recognition will also be a problem with extremely short probes. At the extreme of long probe length, each entire probe sequence is on a different position of a substrate. However, a problem arises from the number of possible sequences, which goes up dramatically with the length of the sequence. Also, the specificity of recognition begins to decrease as,the contribution to binding by any particular subunit may become sufficiently low that the system fails to distinguish the fidelity of recognition. Mismatched hybridization may be a problem with the polynucleotide sequencing applications, though the fingerprinting and mapping applications may not be so strict in their fidelity requirements. As indicated above, a thirty position binary sequence has over a million possible sequences, a number which starts to become unreasonably large in its required number of different sequences, even though the target length is still very short. Preparing a substrate with all sequence possibilities for a long target may be extremely difficult due to the many different oligomers which must be synthesized. [0205]
  • The above example illustrates how a long target sequence may be reconstructed with a reasonably small number of shorter subsequences. Since the present day resolution of the regions of the substrate having defined oligomer probes attached to the substrate approaches about 10 microns by 10 microns for resolvable regions, about 10[0206] 6, or 1 million, positions can be placed on a one centimeter square substrate. However, high resolution systems may have particular disadvantages which may be outweighed using the lower density substrate matrix pattern. For this reason, a sufficiently large number of probe sequences can be utilized so that any given target sequence may be determined by hybridization to a relatively small number of probes.
  • A second complication relates to convergence of sequences to a single subsequence. This will occur when a particular subsequence is repeated in the target sequence. This problem can be addressed in at least two different ways. The first, and simpler way, is to separate the repeat sequences onto two different targets. Thus, each single target will not have the repeated sequence and can be analyzed to its end. This solution, however, complicates the analysis by requiring that some means for cutting at a site between the repeats can be located. Typically a careful sequencer would want to have two intermediate cut points so that the intermediate region can also be sequenced in both directions across each of the cut points. This problem is inherent in the hybridization method for sequencing but can be minimized by using a longer known probe sequence so that the frequency of probe repeats is decreased. [0207]
  • Knowing the sequence of flanking sequences of the repeat will simplify the use of polymerase chain reaction (PCR) or a similar technique to further definitively determine the sequence between sequence repeats. Probes can be made to hybridize to those known sequences adjacent the repeat sequences, thereby producing new target sequences for analysis. See, e.g., Innis et al. (eds.) (1990) [0208] PCR Protocols: A Guide to Methods and Applications, Academic Press; and methods for synthesis of oligonucleotide probes, see, e.g., Gait (1984) Oligonucleotide Synthesis: A Practical Approach, IRL Press, Oxford.
  • Other means for dealing with convergence problems include using particular longer probes, and using degeneracy reducing analogues, see, e.g., Macevicz, S. (1990) PCT publication number WO 90/04652, which is hereby incorporated herein by reference. By use of stretches of the degeneracy reducing analogues with other probes in particular combinations, the number of probes necessary to fully saturate the possible oligomer probes is decreased. For example, with a stretch of 12-mers having the central 4-mer of degenerate nucleotides, in combination with all of the possible 8-mers, the collection numbers twice the number of possible 8-mers, e.g. 65, 536+65,536=131,072, but the population provides screening equivalent to all possible 12-mers. [0209]
  • By way of further explanation, all possible oligonucleotide 8-mers may be depicted in the fashion: [0210]
  • N1-N2-N3-N4-N5-N6-N7-N8, [0211]
  • in which there are 4[0212] 6=65,536 possible 8-mers. As described in Ser. No. 07/624,120, now abandoned, producing all possible 8-mers requires 4×8=32 chemical binary synthesis steps to produce the entire matrix pattern of 65,536 8-mer possibilities. By incorporating degeneracy reducing nucleotides, D's, which hybridize nonselectively to any corresponding complementary nucleotide, new oligonucleotides 12-mers can be made in the fashion:
  • N1-N2-N3-N4-D-D-D-D-N5-N6-N7-N8, [0213]
  • in which there are again, as above, only 4[0214] 6=65,536 possible “12-mers”, which in reality only have 8 different nucleotides.
  • However, it can be seen that each possible 12-mer probe could be represented by a group of the two 8-mer types. Moreover, repeats of less than 12 nucleotides would not converge, or cause repeat problems in the analysis. Thus, instead of requiring a collection of probes corresponding to all 12-mers, or 4[0215] 12=16,777,216 different 12-mers, the same information can be derived by making 2 sets of “8-mers” consisting of the typical 8-mer collection of 48=65,536 and the “12-mer” set with the degeneracy reducing analogues, also requiring making 48=65,536. The combination of the two sets, requires making 65,536+65,536=131,072 different molecules, but giving the information of 16,777,216 molecules. Thus, incorporating the degeneracy reducing analogue decreases the number of molecules necessary to get 12-mer resolution by a factor of about 128-fold.
  • C. Non-polynucleotide Embodiments [0216]
  • The above example is directed towards a polynucleotide embodiment. This application is relatively easily achieved because the specific reagents will typically be complementary oligonucleotides, although in certain embodiments other specific reagents may be desired. For example, there may be circumstances where other than complementary base pairing will be utilized. The polynucleotide targets, will usually be single strand, but may be double or triple stranded in various applications. However, a triple stranded specific interaction might be sometimes desired, or a protein or other specific binding molecule may be utilized. For example, various promoter or DNA sequence specific binding proteins might be used, including, e.g., restriction enzyme binding domains, other binding domains, and antibodies. Thus, specific recognition reagents besides oligonucleotides may be utilized. [0217]
  • For other polymer targets, the specific reagents will often be polypeptides. These polypeptides may be protein binding domains from enzymes or other proteins which display specificity for binding. Usually an antibody molecule may be used, and monoclonal antibodies may be particularly desired. Classical methods may be applied for preparing antibodies, see, e.g., Harlow and Lane (1988) [0218] Antibodies: A Laboratory Manual Cold Spring Harbor Press, New York; and Goding (1986) Monoclonal Antibodies: Principles and Practice (2d Ed.) Academic Press, San Diego. Other suitable techniques for in vitro exposure of lymphocytes to the antigens or selection of libraries of antibody binding sites are described, e.g., in Huse et al. (1989) Science 246:1275-1281; and Ward et al. 91989) Nature 341:544-546, each of which is hereby incorporated herein by reference. Unusual antibody production methods are also described, e.g., in Hendricks et al. (1989) BioTechnology 7:1271-1274; and Hiatt et al. (1989) Nature 342:76-78, each of which is hereby incorporated herein by reference. Other molecules which may exhibit specific binding interaction may be useful for attachment to a VLSIPS substrate by various methods, including the caged biotin methods, see, e.g., Ser. No. 07/435,316, now abandoned, and Barrett et al. (1993) U.S. Pat. No. 5,252,743.
  • The antibody specific reagents should be particularly useful for the polypeptide, carbohydrate, and synthetic polymer applications. Individual specific reagents might be generated by an automated process to generate the number of reagents necessary to advantageously use the high density positional matrix pattern. In an alternative approach, a plurality of hybridoma cells may be screened for their ability to bind to a VLSIPS matrix possessing the desired sequences whose binding specificity is desired. Each cell might be individually grown up and its binding specificity determined by VLSIPS apparatus and technology. An alternative strategy would be to expose the same VLSIPS matrix to a polyclonal serum of high titer. By a successively large volume of serum and different animals, each region of the VLSIPS substrate would have attached to it a substantial number of antibody molecules with specificity of binding. The substrate, with non-covalently bound antibodies could be derivatized and the antibodies transferred to an adjacent second substrate in the matrix pattern in which the antibody molecules had attached to the first matrix. If the sensitivity of detection of binding interaction is sufficiently high, such a low efficiency transfer of antibody molecules may produce a sufficiently high signal to be useful for many purposes, including the sequencing applications. [0219]
  • In another embodiment, capillary forces may be used to transfer the selected reagents to a new matrix, to which the reagents would be positionally attached in the pattern of the recognized sequences. Or, the reagents could be transversely electrophoresed, magnetically transferred, or otherwise transported to a new substrate in their retained positional pattern. [0220]
  • III. Polynucleotide Sequencing [0221]
  • In principle, the making of a substrate having a positionally defined matrix pattern of all possible oligonucleotides of a given length involves a conceptually simple method of synthesizing each and every different possible oligonucleotide, and affixing them to a definable position. Oligonucleotide synthesis is presently mechanized and enabled by current technology, see, e.g., Ser. No. 07/362,901, now abandoned; Pirrung et al. (1992) U.S. Pat. No. 5,143,854; and instruments supplied by Applied Biosystems, Foster City, Calif. [0222]
  • A. Preparation of Substrate Matrix [0223]
  • The production of the collection of specific oligonucleotides used in polynucleotide sequencing may be produced in at least two different ways. Present technology certainly allows production of ten nucleotide oligomers on a solid phase or other synthesizing system. See, e.g., instrumentation provided by Applied Biosystems, Foster City, Calif. Although a single oligonucleotide can be relatively easily made, a large collection of them would typically require a fairly large amount of time and investment. For example, there are 4[0224] 10=1,048,576 possible ten nucleotide oligomers. Present technology allows making each and every one of them in a separate purified form though such might be costly and laborious.
  • Once the desired repertoire of possible oligomer sequences of a given length have been synthesized, this collection of reagents may be individually positionally attached to a substrate, thereby allowing a batchwise hybridization step. Present technology also would allow the possibility of attaching each and every one of these 10-mers to a separate specific position on a solid matrix. This attachment could be automated in any of a number of ways, particularly through the use of a caged biotin type linking. This would produce a matrix having each of different possible 10-mers. [0225]
  • A batchwise hybridization is much preferred because of its reproducibility and simplicity. An automated process of attaching various reagents to positionally defined sites on a substrate is provided in Pirrung et al. (1992) U.S. Pat. No. 5,143,854; Ser. No. 07/624,120, now abandoned; and Barrett et al. (1993) U.S. Pat. No. 5,252,743; each of which is hereby incorporated herein by reference. [0226]
  • Instead of separate synthesis of each oligonucleotide, these oligonucleotides are conveniently synthesized in parallel by sequential synthetic processes on a defined matrix pattern as provided in Pirrung et al. (1992) U.S. Pat. No. 5,143,854; and Ser. No. 07/624,120, now abandoned, which are incorporated herein by reference. Here, the oligonucleotides are synthesized stepwise on a substrate at positionally separate and defined positions. Use of photosensitive blocking reagents allows for defined sequences of synthetic steps over the surface of a matrix pattern. By use of the binary masking strategy, the surface of the substrate can be positioned to generate a desired pattern of regions, each having a defined sequence oligonucleotide synthesized and immobilized thereto. [0227]
  • Although the prior art technology can be used to generate the desired repertoire of oligonucleotide probes, an efficient and cost effective means would be to use the VLSIPS technology described in Pirrung et al. (1992) U.S. Pat. No. 5,143,854 and Ser. No. 07/624,120, now abandoned. In this embodiment, the photosensitive reagents involved in the production of such a matrix are described below. [0228]
  • The regions for synthesis may be very small, usually less than about 100 μm×100 μm, more usually less than about 50 μm×50 μm. The photolithography technology allows synthetic regions of less than about 10 μm×10 μm, about 3 μm×3 μm, or less. The detection also may detect such sized regions, though larger areas are more easily and reliably measured. [0229]
  • At a size of about 30 microns by 30 microns, one million regions would take about 11 centimeters square or a single wafer of about 4 centimeters by 4 centimeters. Thus the present technology provides for making a single matrix of that size having all one million plus possible oligonucleotides. Region size is sufficiently small to correspond to densities of at least about 5 regions/cm[0230] 2, 20 regions/cm2, 50 regions/cm2, 100 regions/cm2, and greater, including 300 regions/cm2, 1000 regions/cm2, 3K regions/cm2, 10K regions/cm2, 30K regions/cm2, 100K regions/cm2, 300K regions/cm2 or more, even in excess of one million regions/cm2.
  • Although the pattern of the regions which contain specific sequences is theoretically not important, for practical reasons certain patterns will be preferred in synthesizing the oligonucleotides. The application of binary masking algorithms for generating the pattern of known oligonucleotide probes is described in related Ser. No. 07/624,120, now abandoned, which was filed simultaneously with this application. By use of these binary masks, a highly efficient means is provided for producing the substrate with the desired matrix pattern of different sequences. Although the binary masking strategy allows for the synthesis of all lengths of polymers, the strategy may be easily modified to provide only polymers of a given length. This is achieved by omitting steps where a subunit is not attached. [0231]
  • The strategy for generating a specific pattern may take any of a number of different approaches. These approaches are well described in related application Ser. No. 07/624,120, now abandoned, and include a number of binary masking approaches which will not be exhaustively discussed herein. However, the binary masking and binary synthesis approaches provide a maximum of diversity with a minimum number of actual synthetic steps. [0232]
  • The length of oligonucleotides used in sequencing applications will be selected on criteria determined to some extent by the practical limits discussed above. For example, if probes are made as oligonucleotides, there will be 65,536 possible eight nucleotide sequences. If a nine subunit oligonucleotide is selected, there are 262,144 possible permeations of sequences. If a ten-mer oligonucleotide is selected, there are 1,048,576 possible permeations of sequences. As the number gets larger, the required number of positionally defined subunits necessary to saturate the possibilities also increases. With respect to hybridization conditions, the length of the matching necessary to confer stability of the conditions selected can be compensated for. See, e.g., Kanehisa, M. (1984) [0233] Nuc. Acids Res. 12:203-213, which is hereby incorporated herein by reference.
  • Although not described in detail here, but below for oligonucleotide probes, the VLSIPS technology would typically use a photosensitive protective group on an oligonucleotide. Sample oligonucleotides are shown in FIG. 1. In particular, the photoprotective group on the nucleotide molecules may be selected from a wide variety of positive light reactive groups preferably including nitro aromatic compounds such as o-nitro-benzyl derivatives or benzylsulfonyl. See, e.g., Gait (1984) [0234] Oligonucleotide Synthesis: A Practical Approach, IRL Press, oxford, which is hereby incorporated herein by reference. In a preferred embodiment, 6-nitro-veratryl oxycarbony (NVOC), 2-nitrobenzyl oxycarbonyl (NBOC), or α,α-dimethyl-dimethoxybenzyl oxycarbonyl (DEZ) is used. Photoremovable protective groups are described in, e.g., Patchornik (1970) J. Amer. Chem. Soc. 92:6333-6335; and Amit et al. (1974) J. Orcanic Chem. 39:192-196; each of which is hereby incorporated herein by reference.
  • A preferred linker for attaching the oligonucleotide to a silicon matrix is illustrated in FIG. 2. A more detailed description is provided below. A photosensitive blocked nucleotide may be attached to specific locations of unblocked prior cycles of attachments on the substrate and can be successively built up to the correct length oligonucleotide probe. [0235]
  • It should be noted that multiple substrates may be simultaneously exposed to a single target sequence where each substrate is a duplicate of one another or where, in combination, multiple substrates together provide the complete or desired subset of possible subsequences. This provides the opportunity to overcome a limitation of the density of positions on a single substrate by using multiple substrates. In the extreme case, each probe might be attached to a single bead or substrate and the beads sorted by whether there is a binding interaction. Those beads which do bind might be encoded to indicate the subsequence specificity of reagents attached thereto. [0236]
  • Then, the target may be bound to the whole collection of beads and those beads that have appropriate specific reagents on them will bind to the target. Then a sorting system may be utilized to sort those beads that actually bind the target from those that do not. This may be accomplished by presently available cell sorting devices or a similar apparatus. After the relatively small number of beads which have bound the target have been collected, the encoding scheme may be read off to determine the specificity of the reagent on the bead. An encoding system may include a magnetic system, a shape encoding system, a color encoding system, or a combination of any of these, or any other encoding system. Once again, with the collection of specific interactions that have occurred, the binding may be analyzed for sequence information, fingerprint information, or mapping information. [0237]
  • The parameters of polynucleotide sizes of both the probes and target sequences are determined by the applications and other circumstances. The length of the oligonucleotide probes used will depend in part upon the limitations of the VLSIPS technology to provide the number of desired probes. For example, in an absolute sequencing application, it is often useful to have virtually all of the possible oligonucleotides of a given length. As indicated above, there are 65,536 8-mers, 262,144 9-mers, 1,048,576 10-mers, 4,194,304 11-mers, etc. As the length of the oligomer increases the number of different probes which must be synthesized also increases at a rate of a factor of 4 for every additional nucleotide. Eventually the size of the matrix and the limitations in the resolution of regions in the matrix will reach the point where an increase in number of probes becomes disadvantageous. However, this sequencing procedure requires that the system be able to distinguish, by appropriate selection of hybridization and washing conditions, between binding of absolute fidelity and binding of complementary sequences containing mismatches. On the other hand, if the fidelity is unnecessary, this discrimination is also unnecessary and a significantly longer probe may be used. Significantly longer probes would typically be useful in fingerprinting or mapping applications. [0238]
  • The length of the probe is selected for a length that will allow the probe to bind with specificity to possible targets. The hybridization conditions are also very important in that they will determine how closely the homology of complementary binding will be detected. In fact, a single target may be evaluated at a number of different conditions to determine its spectrum of specificity for binding particular probes. This may find use in a number of other applications besides the polynucleotide sequencing fingerprinting or mapping. For example, it will be desired to determine the spectrum of binding affinities and specificities of cell surface antigens with binding by particular antibodies immobilized on the substrate surface, particularly under different interaction conditions. In a related fashion, different regions with reagents having differing affinities or levels of specificity may allow such a spectrum to be defined using a single incubation, where various regions, at a given hybridization condition, show the binding affinity. For example, fingerprint probes of various lengths, or with specific defined non-matches may be used. Unnatural nucleotides or nucleotides exhibiting modified specificity of complementary binding are described in greater detail in Macevicz (1990) PCT pub. No. WO 90/04652; and see the section on modified nucleotides in the Sigma Chemical Company catalogue. [0239]
  • B. Labeling Target Nucleotide [0240]
  • The label used to detect the target sequences will be determined, in part, by the detection methods being applied. Thus, the labeling method and label used are selected in combination with the actual detecting systems being used. [0241]
  • Once a particular label has been selected, appropriate labeling protocols will be applied, as described below for specific embodiments. Standard labeling protocols for nucleic acids are described, e.g., in Sambrook et al.; Kambara, H. et al. (1988) [0242] BioTechnology 6:816-821; Smith, L. et al. (1985) Nuc. Acids Res. 13:2399-2412; for polypeptides, see, e.g., Allen G. (1989) Sequencing of Proteins and Peptides, Elsevier, New York, especially chapter 5, and Greenstein and Winitz (1961) Chemistry of the Amino Acids, Wiley and Sons, New York. carbohydrate labeling is described, e.g., in Chaplin and Kennedy (1986) Carbohydrate Analysis: A Practical Approach, IRL Press, Oxford. Labeling of other polymers will be performed by methods applicable to them as recognized by a person having ordinary skill in manipulating the corresponding polymer.
  • In some embodiments, the target need not actually be labeled if a means for detecting where interaction takes place is available. As described below, for a nucleic acid embodiment, such may be provided by an intercalating dye which intercalates only into double stranded segments, e.g., where interaction occurs. See, e.g., Sheldon et al. U.S. Pat. No. 4,582,789. [0243]
  • In many uses, the target sequence will be absolutely homogeneous, both with respect to the total sequence and with respect to the ends of each molecule. Homogeneity with respect to sequence is important to avoid ambiguity. It is preferable that the target sequences of interest not be contaminated with a significant amount of labeled contaminating sequences. The extent of allowable contamination will depend on the sensitivity of the detection system and the inherent signal to noise of the system. Homogeneous contamination sequences will be particularly disruptive of the sequencing procedure. [0244]
  • However, although the target polynucleotide must have a unique sequence, the target molecules need not have identical ends. In fact, the homogeneous target molecule preparation may be randomly sheared to increase the numerical number of molecules. Since the total information content remains the same, the shearing results only in a higher number of distinct sequences which may be labeled and bind to the probe. This fragmentation may give a vastly superior signal relative to a preparation of the target molecules having homogeneous ends. The signal for the hybridization is likely to be dependent on the numerical frequency of the target-probe interactions. If a sequence is individually found on a larger number of separate molecules a better signal will result. In fact, shearing a homogeneous preparation of the target may often be preferred before the labeling procedure is performed, thereby producing a large number of labeling groups associated with each subsequence. [0245]
  • C. Hybridization Conditions [0246]
  • The hybridization conditions between probe and target should be selected such that the specific recognition interaction, i.e., hybridization, of the two molecules is both sufficiently specific and sufficiently stable. See, e.g., Hames and Higgins (1985) [0247] Nucleic Acid Hybridisation: A practical Approach, IRL Press, Oxford. These conditions will be dependent both on the specific sequence and often on the guanine and cytosine (GC) content of the complementary hybrid strands. The conditions may often be selected to be universally equally stable independent of the specific sequences involved. This typically will make use of a reagent such as an alkylammonium buffer. See, Wood et al. (1985) “Base Composition-independent Hybridization in Tetramethylammonium Chloride: A Method for Oligonucleotide Screening of Highly Complex Gene Libraries,” Proc. Natl. Acad. Sci. USA, 82:1585-1588; and Krupov et al. (1989) “An Oligonucleotide Hybridization Approach to DNA Sequencing,” FEBS Letters, 256:118-122; each of which is hereby incorporated herein by reference. An alkylammonium buffer tends to minimize differences in hybridization rate and stability due to GC content. By virtue of the fact that sequences then hybridize with approximately equal affinity and stability, there is relatively little bias in strength or kinetics of binding for particular sequences. Temperature and salt conditions along with other buffer parameters should be selected such that the kinetics of renaturation should be essentially independent of the specific target subsequence or oligonucleotide probe involved. In order to ensure this, the hybridization reactions will usually be performed in a single incubation of all the substrate matrices together exposed to the identical same target probe solution under the same conditions.
  • Alternatively, various substrates may be individually treated differently. Different substrates may be produced, each having reagents which bind to target subsequences with substantially identical stabilities and kinetics of hybridization. For example, all of the high GC content probes could be synthesized on a single substrate which is treated accordingly. In this embodiment, the arylammonium buffers could be unnecessary. Each substrate is then treated in a manner such that the collection of substrates show essentially uniform binding and the hybridization data of target binding to the individual substrate matrix is combined with the data from other substrates to derive the necessary subsequence binding information. The hybridization conditions will usually be selected to be sufficiently specific such that the fidelity of base matching will be properly discriminated. Of course, control hybridizations should be included to determine the stringency and kinetics of hybridization. [0248]
  • D. Detection: VLSIPS™ Technology Scanning [0249]
  • The next step of the sequencing process by hybridization involves labeling of target polynucleotide molecules. A quickly and easily detectable signal is preferred. The VLSIPS™ Technology apparatus is designed to easily detect a fluorescent label, so fluorescent tagging of the target sequence is preferred. Other suitable labels include heavy metal labels, magnetic probes, chromogenic labels (e.g., phosphorescent labels, dyes, and fluorophores) spectroscopic labels, enzyme linked labels, radioactive labels, and labeled binding proteins. Additional labels are described in U.S. Pat. No. 4,366,241, which is incorporated herein by reference. [0250]
  • The detection methods used to determine where hybridization has taken place will typically depend upon the label selected above. Thus, for a fluorescent label a fluorescent detection step will typically be used. Pirrung et al. (1992) U.S. Pat. No. 5,143,854 and Ser. No. 07/624,120, now abandoned, describe apparatus and mechanisms for scanning a substrate matrix using fluorescence detection, but a similar apparatus is adaptable for other optically detectable labels. [0251]
  • The detection method provides a positional localization of the region where hybridization has taken place. However, the position is correlated with the specific sequence of the probe since the probe has specifically been attached or synthesized at a defined substrate matrix position. Having collected all of the data indicating the subsequences present in the target sequence, this data may be aligned by overlap to reconstruct the entire sequence of the target, as illustrated above. [0252]
  • It is also possible to dispense with actual labeling if some means for detecting the positions of interaction between the sequence specific reagent and the target molecule are available. This may take the form of an additional reagent which can indicate the sites either of interaction, or the sites of lack of interaction, e.g., a negative label. For the nucleic acid embodiments, locations of double strand interaction may be detected by the incorporation of intercalating dyes, or other reagents such as antibody or other reagents that recognize helix formation, see, e.g., Sheldon, et al. (1986) U.S. Pat. No. 4,582,789, which is hereby incorporated herein by reference. [0253]
  • E. Analysis [0254]
  • Although the reconstruction can be performed manually as illustrated above, a computer program will typically be used to perform the overlap analysis. A program may be written and run on any of a large number of different computer hardware systems. The variety of operating systems and languages useable will be recognized by a computer software engineer. Various different languages may be used, e.g., BASIC; C; PASCAL; etc. A simple flow chart of data analysis is illustrated in FIG. 1. [0255]
  • F. Substrate Reuse [0256]
  • Finally, after a particular sequence has been hybridized and the pattern of hybridization analyzed, the matrix substrate should be reusable and readily prepared for exposure to a second or subsequent target polynucleotides. In order to do so, the hybrid duplexes are disrupted and the matrix treated in a way which removes all traces of the original target. The matrix may be treated with various detergents or solvents to which the substrate, the oligonucleotide probes, and the linkages to the substrate are inert. This treatment may include an elevated temperature treatment, treatment with organic or inorganic solvents, modifications in pH, and other means for disrupting specific interaction. Thereafter, a second target may actually be applied to the recycled matrix and analyzed as before. [0257]
  • G. Non-Polynucleotide Aspects [0258]
  • Although the sequencing, fingerprinting, and mapping functions will make use of the natural sequence recognition property of complementary nucleotide sequences, the non-polynucleotide sequences typically require other sequence recognition reagents. These reagents will take the form, typically, of proteins exhibiting binding specificity, e.g., enzyme binding sites or antibody binding sites. [0259]
  • Enzyme binding sites may be derived from promoter proteins, restriction enzymes, and the like. See, e.g., Stryer, L. (1988) [0260] Biochemistry, W. H. Freeman, Palo Alto. Antibodies will typically be produced using standard procedures, see, e.g., Harlow and Lane (1988) Antibodies: A Laboratory Manual, Cold Spring Harbor Press, New York; and Goding (1986) Monoclonal Antibodies: Principles and Practice, (2d Ed.) Academic Press, San Diego.
  • Typically, an antigen, or collection of antigens are presented to an immune system. This may take the form of synthesized short polymers produced by the VLSIPS technology, or by the other synthetic means, or from isolation of natural products. For example, antigen for the polypeptides may be made by the VLSIPS technology, by standard peptide synthesis, by isolation of natural proteins with or without degradation to shorter segments, or by expression of a collection of short nucleic acids of random or defined sequences. See, e.g., Tuerk and Gold (1990) [0261] Science 249:505-510, for generation of a collection of randomly mutagenized oligonucleotides useful for expression.
  • The antigen or collection is presented to an appropriate immune system, e.g., to a whole animal as in a standard immunization protocol, or to a collection of immune cells or equivalent. In particular, see Ward et al. (1989) [0262] Nature 341:544-546; and Huse et al. (1989) Science 246:1275-1281, each of which is hereby incorporated herein by reference.
  • A large diversity of antibodies will be generated, some of which have specificities for the desired sequences. Antibodies may be purified having the desired sequence specificities by isolating the cells producing them. For example, a VLSIPS substrate with the desired antigens synthesized thereon may be used to isolate cells with cell surface reagents which recognize the antigens. The VLSIPS substrate may be used as an affinity reagent to select and recover the appropriate cells. Antibodies from those cells may be attached to a substrate using the caged biotin methodology, or by attaching a targeting molecule, e.g., an oligonucleotide. Alternatively, the supernatants from antibody producing cells can be easily assayed using a VLSIPS substrate to identify the cells producing the appropriate antibodies. [0263]
  • Although cells may be isolated, specific antibody molecules which perform the sequence recognition will also be sufficient. Preferably populations of antibody with a known specificity can be isolated. Supernatants from a large population of producing cells may be passed over a VLSIPS substrate to bind to the desired antigens attached to the substrate. When a sufficient density of antibody molecules are attached, they may be removed by an automated process, preferably as antibody populations exhibiting specificity of binding. [0264]
  • In one particular embodiment, a VLSIPS substrate, e.g., with a large plurality of fingerprint antigens attached thereto, is used to isolate antibodies from a supernatant of a population of cells producing antibodies to the antigens. Using the substrate as an affinity reagent, the antibodies will attach to the appropriate positionally defined antigens. The antibodies may be carefully removed therefrom, preferably by an automated system which retains their homogeneous specificities. The isolated antibodies can be attached to a new substrate in a positionally defined matrix pattern. [0265]
  • In a further embodiment, these spatially separated antibodies may be isolated using a specific targeting method for isolation. In this embodiment, a linker molecule which attaches to a particular portion of the antibody, preferably away from the binding site, can be attached to the antibodies. Various reagents will be used, including staphylococcus protein A or antibodies which bind to domains remote from the binding site. Alternatively, the antibodies in the population, before affinity purification, may be derivatized with an appropriate reagent compatible with new VLSIPS synthesis. A preferred reagent is a nucleotide which can serve as a linker to synthetic VLSIPS steps for synthesizing a specific sequence thereon. Then, by successive VLSIPS cycles, each of the antibodies attached to the defined antigen regions can have a defined oligonucleotide synthesized thereon and corresponding in area to the region of the substrate having each antigen attached. These defined oligonucleotides will be useful as targeting reagents to attach those antibodies possessing the same target sequence specificity at defined positions on a new substrate, by virtue of having bound to the antigen region, to a new VLSIPS substrate having the complementary target oligonucleotides positionally located on it. In this fashion, a VLSIPS substrate having the desired antigens attached thereto can be used to generate a second VLSIPS substrate with positionally defined reagents which recognize those antigens. [0266]
  • The selected antigens will typically be selected to be those which define particular functionalities or properties, so as to be useful for fingerprinting and other uses. They will also be useful for mapping and sequencing embodiments. [0267]
  • IV. Fingerprinting [0268]
  • A. General [0269]
  • Many of the procedures and techniques used in the polynucleotide sequencing section are also appropriate for fingerprinting applications. See, e.g., Poustka, et al. (1986) [0270] Cold Spring Harbor Symposia on Ouant. Biol., vol. LI, 131-139, Cold Spring Harbor Press, New York; which is hereby incorporated herein by reference. The fingerprinting method provided herein is based, in part, upon the ability to positionally localize a large number of different specific probes onto a single substrate. This high density matrix pattern provides the ability to screen for, or detect, a very large number of different sequences simultaneously. In fact, depending upon the hybridization conditions, fingerprinting to the resolution of virtually absolute matching of sequence is possible thereby approaching an absolute sequencing embodiment. And the sequencing embodiment is very useful in identifying the probes useful in further fingerprinting uses. For example, characteristic features of genetic sequences will be identified as being diagnostic of the entire sequence. However, in most embodiments, longer probe and target will be used, and for which slight mismatching may not need to be resolved.
  • B. Preparation of Substrate Matrix [0271]
  • A collection of specific probes may be produced by either of the methods described above in the section on sequencing. Specific oligonucleotide probes of desired lengths may be individually synthesized on a standard oligonucleotide synthesizer. The length of these probes is limited only by the ability of the synthesizer to continue to accurately synthesize a molecule. Oligonucleotides or sequence fragments may also be isolated from natural sources. Biological amplification methods may be coupled with synthetic synthesizing procedures such as, e.g., polymerase chain reaction. [0272]
  • In one embodiment, the individually isolated probes may be attached to the matrix at defined positions. These probe reagents may be attached by an automated process making use of the caged biotin methodology described in Ser. No. 07/612,671, or using photochemical reagents, see, e.g., Dattagupta et al. (1985) U.S. Pat. No. 4,542,102 and (1987) U.S. Pat. No. 4,713,326. Each individually purified reagent can be attached individually at specific locations on a substrate. [0273]
  • In another embodiment, the VLSIPS synthesizing technique may be used to synthesize the desired probes at specific positions on a substrate. The probes may be synthesized by successively adding appropriate monomer subunits, e.g., nucleotides, to generate the desired sequences. [0274]
  • In another embodiment, a relatively short specific oligonucleotide is used which serves as a targeting reagent for positionally directing the sequence recognition reagent. For example, the sequence specific reagents having a separate additional sequence recognition segment (usually of a different polymer from the target sequence) can be directed to target oligonucleotides attached to the substrate. By use of non-natural targeting reagents, e.g., unusual nucleotide analogues which pair with other unnatural nucleotide analogues and which do not interfere with natural nucleotide interactions, the natural and non-natural portions can coexist on the same molecule without interfering with their individual functionalities. This can combine both a synthetic and biological production system analogous to the technique for targeting monoclonal antibodies to locations on a VLSIPS substrate at defined positions. Unnatural optical isomers of nucleotides may be useful unnatural reagents subject to similar chemistry, but incapable of interfering with the natural biological polymers. See also, Ser. No. 07/626,730, which is hereby incorporated herein by reference. [0275]
  • After the separate substrate attached reagents are attached to the targeting segment, the two are crosslinked, thereby permanently attaching them to the substrate. Suitable crosslinking reagents are known, see, e.g., Dattagupta et al. (1985) U.S. Pat. No. 4,542,102 and (1987) “Coupling of nucleic acids to solid support by photochemical methods,” U.S. Pat. No. 4,713,326, each of which is hereby incorporated herein by reference. Similar linkages for attachment of proteins to a solid substrate are provided, e.g., in Merrifield (1986) [0276] Science 232:341-347, which is hereby incorporated herein by reference.
  • C. Labelinc Target Nucleotides [0277]
  • The labeling procedures used in the sequencing embodiments will also be applicable in the fingerprinting embodiments. However, since the fingerprinting embodiments often will involve relatively large target molecules and relatively short oligonucleotide probes, the amount of signal necessary to incorporate into the target sequence may be less critical than in the sequencing applications. For example, a relatively long target with a relatively small number of labels per molecule may be easily amplified or detected because of the relatively large target molecule size. [0278]
  • In various embodiments, it may be desired to cleave the target into smaller segments as in the sequencing embodiments. The labeling procedures and cleavage techniques described in the sequencing embodiments would usually also be applicable here. [0279]
  • D. Hybridization Conditions [0280]
  • The hybridization conditions used in fingerprinting embodiments will typically be less critical than for the sequencing embodiments. The reason is that the amount of mismatching which may be useful in providing the fingerprinting information would typically be far greater than that necessary in sequencing uses. For example, Southern hybridizations do not typically distinguish between slightly mismatched sequences. Under these circumstances, important and valuable information may be arrived at with less stringent hybridization conditions while providing valuable fingerprinting information. However, since the entire substrate is typically exposed to the target molecule at one time, the binding affinity of the probes should usually be of approximately comparable levels. For this reason, if oligonucleotide probes are being used, their lengths should be approximately comparable and will be selected to hybridize under conditions which are common for most of the probes on the substrate. Much as in a Southern hybridization, the target and oligonucleotide probes are of lengths typically greater than about 25 nucleotides. Under appropriate hybridization conditions, e.g., typically higher salt and lower temperature, the probes will hybridize irrespective of imperfect complementarity. In fact, with probes of greater than, e.g., about fifty nucleotides, the difference in stability of different sized probes will be relatively minor. [0281]
  • Typically the fingerprinting is merely for probing similarity or homology. Thus, the stringency of hybridization can usually be decreased to fairly low levels. See, e.g., Wetmur and Davidson (1968) “Kinetics of Renaturation of DNA,” [0282] J. Mol. Biol., 31:349-370; and Kanehisa, M. (1984) Nuc. Acids Res., 12:203-213.
  • E. Detection: VLSIPS™ Technology Scanning [0283]
  • Detection methods will be selected which are appropriate for the selected label. The scanning device need not necessarily be digitized or placed into a specific digital database, though such would most likely be done. For example, the analysis in fingerprinting could be photographic. Where a standardized fingerprint substrate matrix is used, the pattern of hybridizations may be spatially unique and may be compared photographically. In this manner, each sample may have a characteristic pattern of interactions and the likelihood of identical patterns will preferably be such low frequency that the fingerprint pattern indeed becomes a characteristic pattern virtually as unique as an individual's fingertip fingerprint. With a standardized substrate, every individual could be, in theory, uniquely identifiable on the basis of the pattern of hybridizing to the substrate. [0284]
  • Of course, the VLSIPS™ Technology scanning apparatus may also be useful to generate a digitized version of the fingerprint pattern. In this way, the identification pattern can be provided in a linear string of digits. This sequence could also be used for a standardized identification system providing significant useful medical transferability of specific data. In one embodiment, the probes used are selected to be of sufficiently high resolution to measure the antigens of the major histo compatibility complex. It might even be possible to provide transplantation matching data in a linear stream of data. The fingerprinting data may provide a condensed version, or summary, of the linear genetic data, or any other information data base. [0285]
  • F. Analysis [0286]
  • The analysis of the fingerprint will often be much simpler than a total sequence determination. However, there may be particular types of analysis which will be substantially simplified by a selected group of probes. For example, probes which exhibit particular populational heterogeneity may be selected. In this way, analysis may be simplified and practical utility enhanced merely by careful selection of the specific probes and a careful matrix layout of those probes. [0287]
  • G. Substrate Reuse [0288]
  • As with the sequencing application, the fingerprinting usages may also take advantage of the reusability of the substrate. In this way, the interactions can be disrupted, the substrate treated, and the renewed substrate is equivalent to an unused substrate. [0289]
  • H. Non-polynucleotide Aspects [0290]
  • Besides polynucleotide applications, the fingerprinting analysis may be applied to other polymers, especially polypeptides, carbohydrates, and other polymers, both organic and inorganic. Besides using the fingerprinting method for analyzing a particular polymer, the fingerprinting method may be used to characterize various samples. For example, a cell or population of cells may be tested for their expression of specific antigens or their mRNA sequence intent. For example, a T-cell may be classified by virtue of its combination of expressed surface antigens. With specific reagents which interact with these antigens, a cell or a population of cells or a lysed cell may be exposed to a VLSIPS substrate. The biological sample may be classified or characterized by analyzing the pattern of specific interaction. This may be applicable to a cell or tissue type, to the messenger RNA population expressed by a cell to the genetic content of a cell, or to virtually any sample which can be classified and/or identified by its combination of specific molecular properties. [0291]
  • The ability to generate a high density means for screening the presence or absence of specific interactions allows for the possibility of screening for, if not saturating, all of a very large number of possible interactions. This is very powerful in providing the means for testing the combinations of molecular properties which can define a class of samples. For example, a species of organism may be characterized by its DNA sequences, e.g., a genetic fingerprint. By using a fingerprinting method, it may be determined that all members of that species are sufficiently similar in specific sequences that they can be easily identified as being within a particular group. Thus, newly defined classes may be resolved by their similarity in fingerprint patterns. Alternatively, a non-member of that group will fail to share those many identifying characteristics. However, since the technology allows testing of a very large number of specific interactions, it also provides the ability to more finely distinguish between closely related different cells or samples. This will have important applications in diagnosing viral, bacterial, and other pathological on nonpathological infections. [0292]
  • In particular, cell classification may be defined by any of a number of different properties. For example, a cell class may be defined by its DNA sequences contained therein. This allows species identification for parasitic or other infections. For example, the human cell is presumably genetically distinguishable from a monkey cell, but different human cells will share many genetic markers. At higher resolution, each individual human genome will exhibit unique sequences that can define it as a single individual. [0293]
  • Likewise, a developmental stage of a cell type may be definable by its pattern of expression of messenger RNA. For example, in particular stages of cells, high levels of ribosomal RNA are found whereas relatively low levels of other types of messenger RNAs may be found. The high resolution distinguishability provided by this fingerprinting method allows the distinction between cells which have relatively minor differences in its expressed mRNA population. Where a pattern is shown to be characteristic of a stage, a stage may be defined by that particular pattern of messenger RNA expression. [0294]
  • In a similar manner, the antigenic determinants found on a protein may very well define the cell class. For example, immunological T-cells are distinguishable from B-cells because, in part, the cell surface antigens on the cell types are distinguishable. Different T-cell subclasses can be also distinguished from one another by whether they contain particular T-cell antigens. The present invention provides the possibility for high resolution testing of many different interactions simultaneously, and the definition of new cell types will be possible. [0295]
  • The high resolution VLSIPS™ substrate may also be used as a very powerful diagnostic tool to test the combination of presence, of a plurality of different assays from a biological sample. For example, a cancerous condition may be indicated by a combination of various different properties found in the blood. For example, a cancerous condition may be indicated by a combination of expression of various soluble antigens found in the blood along with a high number of various cellular antigens found on lymphocytes and/or particular cell degradation products. With a substrate as provided herein, a large number of different features can be simultaneously performed on a biological sample. In fact, the high resolution of the test will allow more complete characterization of parameters which define particular diseases. Thus, the power of diagnostic tests may be limited by the extent of statistical correlation with a particular condition rather than with the number of antigens or interactions which are tested. The present invention provides the means to generate this large universe of possible reagents and the ability to actually accumulate that correlative data. [0296]
  • In another embodiment, a substrate as provided herein may be used for genetic screening. This would allow for simultaneous screening of thousands of genetic markers. As the density of the matrix is increased, many more molecules can be simultaneously tested. Genetic screening then becomes a simpler method as the present invention provides the ability to screen for thousands, tens of thousands, and hundreds of thousands, even millions of different possible genetic features. However, the number of high correlation genetic markers for conditions numbers only in the hundreds. Again, the possibility for screening a large number of sequences provides the opportunity for generating the data which can provide correlation between sequences and specific conditions or susceptibility. The present invention provides the means to generate extremely valuable correlations useful for the genetic detection of the causative mutation leading to medical conditions. In still another embodiment, the present invention would be applicable to distinguishing two individuals having identical genetic compositions. The antibody population within an individual is dependent both on genetic and historical factors. Each individual experiences a unique exposure to various infectious agents, and the combined antibody expression is partly determined thereby. Thus, individuals may also be fingerprinted by their immunological content, either of actively expressed antibodies, or their immunological memory. Similar sorts of immunological and environmental histories may be useful for fingerprinting, perhaps in combination with other screening properties. In particular, the present invention may be useful for screening allergic reactions or susceptibilities, and a simple IgE specificity test may be useful in determining a spectrum of allergies. [0297]
  • With the definition of new classes of cells, a cell sorter will be used to purify them. Moreover, new markers for defining that class of cells will be identified. For example, where the class is defined by its RNA content, cells may be screened by antisense probes which detect the presence or absence of specific sequences therein. Alternatively, cell lysates may provide information useful in correlating intracellular properties with extracellular markers which indicate functional differences. Using standard cell sorter technology with a fluorescence or labeled antisense probe which recognizes the internal presence of the specific sequences of interest, the cell sorter will be able to isolate a relatively homogeneous population of cells possessing the particular marker. Using successive probes the sorting process should be able to select for cells having a combination of a large number of different markers. [0298]
  • In a non-polynucleotide embodiment, cells may be defined by the presence of other markers. The markers may be carbohydrates, proteins, or other molecules. Thus, a substrate having particular specific reagents, e.g., antibodies, attached to it should be able to identify cells having particular patterns of marker expression. Of course, combinations of these made be utilized and a cell class may be defined by a combination of its expressed mRNA, its carbohydrate expression, its antigens, and other properties. This fingerprinting should be useful in determining the physiological state of a cell or population of cells. [0299]
  • Having defined a cell type whose function or properties are defined by the reagents attachable to a VLSIPS substrate, such as cellular antigens, these structural manifestations of function may be used to sort cells to generate a relatively homogeneous population of that class of cells. Standard cell sorter technology may be applied to purify such a population, see, e.g., Dangl, J. and Herzenberg (1982) “Selection of hybridomas and hybridoma variants using the fluorescence activated cell sorter,” [0300] J. Immunological Methods 52:1-14; and Becton Dickinson, Fluorescence Activated Cell Sorter Division, San Jose, California, and Coulter Diagnostics, Hialeah, Florida.
  • With the fingerprinting method an identification means arises from mosaicism problems in an organism. A mosaic organism is one whose genetic content in different cells is significantly different. Various clonal populations should have similar genetic fingerprints, though different clonal populations may have different genetic contents. See, for example, Suzuki et al. [0301] An Introduction to Genetic Analysis (4th Ed.), Freeman and Co., New York, which is hereby incorporated herein by reference. However, this problem should be a relatively rare problem and could be more carefully evaluated with greater experience using the fingerprinting methods.
  • The invention will also find use in detecting changes, both genetic and antigenic, e.g., in a rapidly “evolving” protozoa infection, or similarly changing organism. [0302]
  • V. Mapping [0303]
  • A. General [0304]
  • The use of the present invention for mapping parallels its use for fingerprinting and sequencing. Where a polymer is a linear molecule, the mapping provides the ability to locate particular segments along the length of the polymer. Branched polymers can be treated as a series of individual linear polymers. The mapping provides the ability to locate, in a relative sense, the order of various subsequences. This may be achieved using at least two different approaches. [0305]
  • The first approach is to take the large sequence and fragment it at specific points. The fragments are then ordered and attached to a solid substrate. For example, the clones resulting from a chromosome walking process may be individually attached to the substrate by methods, e.g., caged biotin techniques, indicated earlier. Segments of unknown map position will be exposed to the substrate and will hybridize to the segment which contains that particular sequence. This procedure allows the rapid determination of a number of different labeled segments, each mapping requiring only a single hybridization step once the substrate is generated. The substrate may be regenerated by removal of the interaction, and the next mapping segment applied. [0306]
  • In an alternative method, a plurality of subsequences can be attached to a substrate. Various short probes may be applied to determine which segments may contain particular overlaps. The theoretical basis and a description of this mapping procedure is contained in, e.g., Evans et al. 1989 “Physical Mapping of Complex Genomes by Cosmid Multiplex Analysis,” [0307] Proc. Natl. Acad. Sci. USA 86:5030-5034, and other references cited above in the Section labeled “Overall Description.” Using this approach, the details of the mapping embodiment are very similar to those used in the fingerprinting embodiment.
  • B. Preparation of Substrate Matrix [0308]
  • The substrate may be generated in either of the methods generally applicable in the sequencing and fingerprinting embodiments. The substrate may be made either synthetically, or by attaching otherwise purified probes or sequences to the matrix. The probes or sequences may be derived either from synthetic or biological means. As indicated above, the solid phase substrate synthetic methods may be utilized to generate a matrix with positionally defined sequences. In the mapping embodiment, the importance of saturation of all possible subsequences of a preselected length is far less important than in the sequencing embodiment, but the length of the probes used may be desired to be much longer. The processes for making a substrate which has longer oligonucleotide probes should not be significantly different from those described for the sequencing embodiments, but the optimization parameters may be modified to comply with the mapping needs. [0309]
  • C. Labeling [0310]
  • The labeling methods will be similar to those applicable in sequencing and fingerprinting embodiments. Again, it may be desirable to fragment the target sequences. [0311]
  • D. Hybridization/Specific Interaction [0312]
  • The specificity of interaction between the targets and probe would typically be closer to those used for fingerprinting embodiments, where homology is more important than absolute distinguishability of high fidelity complementary hybridization. Usually, the hybridization conditions will be such that merely homologous segments will interact and provide a positive signal. Much like the fingerprinting embodiment, it may be useful to measure the extent of homology by successive incubations at higher stringency conditions. Or, a plurality of different probes, each having various levels of homology may be used. In either way, the spectrum of homologies can be measured. [0313]
  • Where non-nucleic acid hybridization is involved, the specific interactions may also be compared in a fingerprint-like manner. The specific reagents may have less specificity, e.g., monoclonal antibodies which recognize a broader spectrum of sequences may be utilized relative to a sequencing embodiment. Again, the specificity of interaction may be measured under various conditions of increasing stringency to determine the spectrum of matching across the specific probes selected, or a number of different stringency reagents may be included to indicate the binding affinity. [0314]
  • E. Detection [0315]
  • The detection methods used in the mapping procedure will be virtually identical to those used in the fingerprinting embodiment. The detection methods will be selected in combination with the labeling methods. [0316]
  • F. Analysis [0317]
  • The analysis of the data in a mapping embodiment will typically be somewhat different from that in fingerprinting. The fingerprinting embodiment will test for the presence or absence of specific or homologous segments. However, in the mapping embodiment, the existence of an interaction is coupled with some indication of the location of the interaction. The interaction is mapped in some manner to the physical polymer sequence. Some means for determining the relative positions of different probes is performed. This may be achieved by synthesis of the substrate in pattern, or may result from analysis of sequences after they have been attached to the substrate. [0318]
  • For example, the probes may be randomly positioned at various locations on the substrate. However, the relative positions of the various reagents in the original polymer may be determined by using short fragments, e.g., individually, as target molecules which determine the proximity of different probes. By an automated system of testing each different short fragment of the original polymer, coupled with proper analysis, it will be possible to determine which probes are adjacent one another on the original target sequence and correlate that with positions on the matrix. In this way, the matrix is useful for determining the relative locations of various new segments in the original target molecule. This sort of analysis is described in Evans, and the related references described above. [0319]
  • G. Substrate Reuse [0320]
  • The substrate should be reusable in the manner described in the fingerprinting section. The substrate is renewed by removal of the specific interactions and is washed and prepared for successive cycles of exposure to new target sequences. [0321]
  • H. Non-polynucleotide Aspects [0322]
  • The mapping procedure may be used on other molecules than polynucleotides. Although hybridization is one type of specific interaction which is clearly useful for use in this mapping embodiment, antibody reagents may also be very useful. In the same way that polypeptide sequencing or other polymers may be sequenced by the reagents and techniques described in the sequencing section and fingerprinting section, the mapping embodiment may also be used similarly. [0323]
  • In another form of mapping, as described above in the fingerprinting section, the developmental map of a cell or biological system may be measured using fingerprinting type technology. Thus, the mapping may be along a temporal dimension rather than along a polymer dimension. The mapping or fingerprinting embodiments may also be used in determining the genetic rearrangements which may be genetically important, as in lymphocyte and B-cell development. In another example, various rearrangements or chromosomal dislocations may be tested by either the fingerprinting or mapping methods. These techniques are similar in many respects and the fingerprinting and mapping embodiments may overlap in many respects. [0324]
  • VI. Additional Screening and Applications [0325]
  • A. Specific Interactions [0326]
  • As originally indicated in the parent filing of VLSIPS™ Technology, the production of a high density plurality of spatially segregated polymers provides the ability to generate a very large universe or repertoire of individually and distinct sequence possibilities. As indicated above, particular oligonucleotides may be synthesized in automated fashion at specific locations on a matrix. In fact, these oligonucleotides may be used to direct other molecules to specific locations by linking specific oligonucleotides to other reagents which are in batch exposed to the matrix and hybridized in a complementary fashion to only those locations where the complementary oligonucleotide has been synthesized on the matrix. This allows for spatially attaching a plurality of different reagents onto the matrix instead of individually attaching each separate reagent at each specific location. Although the caged biotin method allows automated attachment, the speed of the caged biotin attachment process is relatively slow and requires a separate reaction for each reagent being attached. By use of the oligonucleotide method, the specificity of position can be done in an automated and parallel fashion. As each reagent is produced, instead of directly attaching each reagent at each desired position, the reagent may be attached to a specific desired complementary oligonucleotide which will ultimately be specifically directed toward locations on the matrix having a complementary oligonucleotide attached thereat. [0327]
  • In addition, the technology allows screening for specificity of interaction with particular reagents. For example, the oligonucleotide sequence specificity of binding of a potential reagent may be tested by presenting to the reagent all of the possible subsequences available for binding. Although secondary or higher order sequence specific features might not be easily screenable using this technology, it does provide a convenient, simple, quick, and thorough screen of interactions between a reagent and its target recognition sequences. See, e.g., Pfeifer et al. (1989) [0328] Science 246:810-812.
  • For example, the interaction of a promoter protein with its target binding sequence may be tested for many different, or all, possible binding sequences. By testing the strength of interactions under various different conditions, the interaction of the promoter protein with each of the different potential binding sites may be analyzed. The spectrum of strength of interactions with each different potential binding site may provide significant insight into the types of features which are important in determining specificity. [0329]
  • An additional example of a sequence specific interaction between reagents is the testing of binding of a double stranded nucleic acid structure with a single stranded oligonucleotide. Often, a triple stranded structure is produced which has significant aspects of sequence specificity. Testing of such interactions with either sequences comprising only natural nucleotides, or perhaps the testing of nucleotide analogs may be very important in screening for particularly useful diagnostic or therapeutic reagents. See, e.g., Haner and Dervan (1990) [0330] Biochemistry 29:9761-6765, and references therein.
  • B. Seauence Comparisons [0331]
  • Once a gene is sequenced, the present invention provides a means to compare alleles or related sequences to locate and identify differences from the control sequence. This would be extremely useful in further analysis of genetic variability at a specific gene locus. [0332]
  • C. Categorizations [0333]
  • As indicated above in the fingerprinting and mapping embodiments, the present invention is also useful in defining specific stages in the temporal sequence of cells, e.g., development, and the resulting tissues within an organism. For example, the developmental stage of a cell, or population of cells, can be dependent upon the expression of particular messenger RNAs or cellular antigens. The screening procedures provided allow for high resolution definition of new classes of cells. In addition, the temporal development of particular cells will be characterized by the presence or expression of various mRNAs. Means to simultaneously screen a plurality or very large number of different sequences are provided. The combination of different markers made available dramatically increases the ability to distinguish fairly closely related cell types. Other markers may be combined with markers and methods made available herein to define new classifications of biological samples, e.g., based upon new combinations of markers. [0334]
  • The presence or absence of particular marker sequences will be used to define temporal developmental stages. Once the stages are defined, fairly simple methods can be applied to actually purify those particular cells. For example, antisense probes or recognition reagents may be used with a cell sorter to select those cells containing or expressing the critical markers. Alternatively, the expression of those sequences may result in specific antigens which may also be used in defining cell classes and sorting those cells away from others. In this way, for example, it should be possible to select a class of omnipotent immune system cells which are able to completely regenerate a human immune system. Based upon the cellular classes defined by the parameters made available by this technology, purified classes of cells having identifiable differences, structural or functional, are made available. [0335]
  • In an alternative embodiment, a plurality of antigens or specific binding proteins attached to the substrate may be used to define particular cell types. For example, subclasses of T-cells are defined, in part, by the combination of expressed cell surface antigens. The present invention allows for the simultaneous screening of a large plurality of different antigens together. Thus, higher resolution classification of different T-cell subclasses becomes possible and, with the definitions and functional differences which correlate with those antigenic or other parameters, the ability to purify those cell types becomes available. This is applicable not only to T-cells, but also to lymphocyte cells, or even to freely circulating cells. Many of the cells for which this would be most useful will be immobile cells found in particular tissues or organs. Tumor cells will be diagnosed or detected using these fingerprinting techniques. Coupled with a temporal change in structure, developmental classes may also be selected and defined using these technologies. The present invention also provides the ability not only to define new classes of cells based upon functional or structural differences, but it also provides the ability to select or purify populations of cells which share these particular properties. Standard cell sorting procedures using antibody markers may be used to detect extracellular features. Intracellular features would also be detectable by introducing the label reagents into the cell. In particular, antisense DNA or RNA molecules may be introduced into a cell to detect RNA sequences therein. See, e.g., Weintraub (1990) [0336] Scientific American 262:40-46.
  • D. Statistical Correlations [0337]
  • In an additional embodiment, the present invention also allows for the high resolution correlation of medical conditions with various different markers. For example, the presently available technology, when applied to amniocentesis or other genetic screening methods, typically screens for tens of different markers at most. The present invention allows simultaneous screening for tens, hundreds, thousands, tens of thousands, hundreds of thousands, and even millions of different genetic sequences. Thus, applying the fingerprinting methods of the present invention to a sufficiently large population allows detailed statistical analysis to be made, thereby correlating particular medical conditions with particular markers, typically antigenic or genetic. Tumor specific antigens will be identified using the present invention. [0338]
  • Various medical conditions may be correlated against an enormous data base of the sequences within an individual. Genetic propensities and correlations then become available and high resolution genetic predictability and correlation become much more easily performed. With the enormous data base, the reliability of the predictions is also better tested. Particular markers which are partially diagnostic of particular medical conditions or medical susceptibilities will be identified and provide direction in further studies and more careful analysis of the markers involved. Of course, as indicated above in the sequencing embodiment, the present invention will find much use in intense sequencing projects. For example, sequencing of the entire human genome in the human genome project will be greatly simplified and enabled by the present invention. [0339]
  • VI. Formation of Substrate [0340]
  • The substrate is provided with a pattern of specific reagents which are positionally localized on the surface of the substrate. This matrix of positions is defined by the automated system which produces the substrate. The instrument will typically be one similar to that described in Pirrung et al. (1992) U.S. Pat. No. 5,143,854, and Ser. No. 07/624,120, now abandoned. The instrumentation described therein is directly applicable to the applications used here. In particular, the apparatus comprises a substrate, typically a silicon containing substrate, on which positions on the surface may be defined by a coordinate system of positions. These positions can be individually addressed or detected by the VLSIPS™ Technology apparatus. [0341]
  • Typically, the VLSIPS™ Technology apparatus uses optical methods used in semiconductor fabrication applications. In this way, masks may be used to photo-activate positions for attachment or synthesis of specific sequences on the substrate. These manipulations may be automated by the types of apparatus described in Pirrung et al. (1992) U.S. Pat. No. 5,143,854 and Ser. No. 07/624,120, now abandoned. [0342]
  • Selectively removable protecting groups allow creation of well defined areas of substrate surface having differing reactivities. Preferably, the protecting groups are selectively removed from the surface by applying a specific activator, such as electromagnetic radiation of a specific wavelength and intensity. More preferably, the specific activator exposes selected areas of surface to remove the protecting groups in the exposed areas. [0343]
  • Protecting groups of the present invention are used in conjunction with solid phase oligomer syntheses, such as peptide syntheses using natural or unnatural amino acids, nucleotide syntheses using deoxyribonucleic and ribonucleic acids, oligosaccharide syntheses, and the like. In addition to protecting the substrate surface from unwanted reaction, the protecting groups block a reactive end of the monomer to prevent self-polymerization. For instance, attachment of a protecting group to the amino terminus of an activated amino acid, such as the N-hydroxysuccinimide-activated ester of the amino acid prevents the amino terminus of one monomer from reacting with the activated ester portion of another during peptide synthesis. [0344]
  • Alternatively, the protecting group may be attached to the carboxyl group of an amino acid to prevent reaction at this site. Most protecting groups can be attached to either the amino or the carboxyl group of an amino acid, and the nature of the chemical synthesis will dictate which reactive group will require a protecting group. Analogously, attachment of a protecting group to the 5′-hydroxyl group of a nucleoside during synthesis using for example, phosphate-triester coupling chemistry, prevents the 5′-hydroxyl of one nucleoside from reacting with the 3′-activated phosphate-triester of another. [0345]
  • Regardless of the specific use, protecting groups are employed to protect a moiety on a molecule from reacting with another reagent. Protecting groups of the present invention have the following characteristics: they prevent selected reagents from modifying the group to which they are attached; they are stable (that is, they remain attached) to the synthesis reaction conditions; they are removable under conditions that do not adversely affect the remaining structure; and once removed, do not react appreciably with the surface or surface-bound oligomer. The selection of a suitable protecting group will depend, of course, on the chemical nature of the monomer unit and oligomer, as well as the specific reagents they are to protect against. [0346]
  • In a preferred embodiment, the protecting groups will be photoactivatable. The properties and uses of photoreactive protecting compounds have been reviewed. See, McCray et al., [0347] Ann. Rev. of Biophys. and Biophys. Chem. (1989) 18:239-270, which is incorporated herein by reference. Preferably, the photosensitive protecting groups will be removable by radiation in the ultraviolet (UV) or visible portion of the electromagnetic spectrum. More preferably, the protecting groups will be removable by radiation in the near UV or visible portion of the spectrum. In some embodiments, however, activation may be performed by other methods such as localized heating, electron beam lithography, laser pumping, oxidation or reduction with microelectrodes, and the like. Sulfonyl compounds are suitable reactive groups for electron beam lithography. Oxidative or reductive removal is accomplished by exposure of the protecting group to an electric current source, preferably using microelectrodes directed to the predefined regions of the surface which are desired for activation. A more detailed description of these protective groups is provided in Ser. No. 07/624,120, now abandoned, which is hereby incorporated herein by reference.
  • The density of reagents attached to a silicon substrate may be varied by standard procedures. The surface area for attachment of reagents may be increased by modifying the silicon surface. For example, a matte surface may be machined or etched on the substrate to provide more sites for attachment of the particular reagents. Another way to increase the density of reagent binding sites is to increase the derivitization density of the silicon. Standard procedures for achieving this are described, below. [0348]
  • One method to control the derivatization density is to highly derivatize the substrate with photochemical groups at high density. The substrate is then photolyzed for various predetermined times, which photoactivate the groups at a measurable rate, and react them with a capping reagent. By this method, the density of linker groups may be modulated by using a desired time and intensity of photoactivation. [0349]
  • In many applications, the number of different sequences which may be provided may be limited by the density and the size of the substrate on which the matrix pattern is generated. In situations where the density is insufficiently high to allow the screening of the desired number of sequences, multiple substrates may be used to increase the number of sequences tested. Thus, the number of sequences tested may be increased by using a plurality of different substrates. Because the VLSIPS apparatus is almost fully automated, increasing the number of substrates does not lead to a significant increase in the number of manipulations which must be performed by humans. This again leads to greater reproducibility and speed in the handling of these multiple substrates. [0350]
  • A. Instrumentation [0351]
  • The concept of using VLSIPS™ Technology generally allows a pattern or a matrix of reagents to be generated. The procedure for making the pattern is performed by any of a number of different methods. An apparatus and instrumentation useful for generating a high density VLSIPS substrate is described in detail in Pirrung et al. (1992) U.S. Pat. No. 5,143,854 and Ser. No. 07/624,120, now abandoned. [0352]
  • B. Binary Masking [0353]
  • The details of the binary masking are described in an accompanying application filed simultaneously with this, Ser. No. 07/624,120, now abandoned, whose specification is incorporated herein by reference. [0354]
  • For example, the binary masking technique allows for producing a plurality of sequences based on the selection of either of two possibilities at any particular location. By a series of binary masking steps, the binary decision may be the determination, on a particular synthetic cycle, whether or not to add any particular one of the possible subunits. By treating various regions of the matrix pattern in parallel, the binary masking strategy provides the ability to carry out spatially addressable parallel synthesis. [0355]
  • C. Synthetic Methods [0356]
  • The synthetic methods in making a substrate are described in the parent application, Pirrung et al. (1992) U.S. Pat. No. 5,143,854. The construction of the matrix pattern on the substrate will typically be generated by the use of photo-sensitive reagents. By use of photo-lithographic optical methods, particular segments of the substrate can be irradiated with light to activate or deactivate blocking agents, e.g., to protect or deprotect particular chemical groups. By an appropriate sequence of photo-exposure steps at appropriate times with appropriate masks and with appropriate reagents, the substrates can have known polymers synthesized at positionally defined regions on the substrate. Methods for synthesizing various substrates are described in Pirrung et al. (1992) U.S. Pat. No. 5,143,854 and Ser. No. 07/624,120, now abandoned. By a sequential series of these photo-exposure and reaction manipulations, a defined matrix pattern of known sequences may be generated, and is typically referred to as a VLSIPS™ Technology substrate. In the nucleic acid synthesis embodiment, nucleosides used in the synthesis of DNA by photolytic methods will typically be one of the two forms shown below: [0357]
    Figure US20020155491A1-20021024-C00001
  • B=Adenine, Cytosine, Guanine, or Thymine [0358]
  • In I, the photolabile group at the 5′ position is abbreviated UV (nitroveratryl) and in II, the group is abbreviated NVOC (nitroveratryl oxycarbonyl). Although not shown in FIG. C, the bases (adenine, cytosine, and guanine) contain exocyclic NH[0359] 2 groups which must be protected during DNA synthesis. Thymine contains no exocyclic NH2 and therefore requires no protection. The standard protecting groups for these amines are shown below:
    Figure US20020155491A1-20021024-C00002
  • other amides of the general formula [0360]
    Figure US20020155491A1-20021024-C00003
  • where R may be alkyl or aryl have been used. [0361]
  • Another type of protecting group FMOC (9-fluorenyl methoxycarbonyl) is currently being used to protect the exocyclic amines of the three bases: [0362]
    Figure US20020155491A1-20021024-C00004
  • Adenine (A) Cytosine (C) Guanine (G) [0363]
  • The advantage of the FMCC group is that it is removed under mild conditions (dilute organic bases) and can be used for all three bases. The amide protecting groups require more harsh conditions to be removed (NH[0364] 3/MeOH with heat)
  • Nucleosides used as 5′-OH probes, useful in verifying correct VLSIPS synthetic function, include, for example, the following: [0365]
    Figure US20020155491A1-20021024-C00005
  • These compounds are used to detect where on a substrate photolysis has occurred by the attachment of either III or V to the newly generated 5′-OH. In the case of III, after the phosphate attachment is made, the substrate is treated with a dilute base to remove the FMOC group. The resulting amine can be reacted with FITC and the substrate examined by fluorescence microscopy. This indicates the proper generation of a 5′-OH. In the case of compound IV, after the phosphate attachment is made, the substrate is treated with FITC labeled streptavidin and the substrate again may be examined by fluorescence microscopy. Other probes, although not nucleoside based, have included the following: [0366]
    Figure US20020155491A1-20021024-C00006
  • The method of attachment of the first nucleoside to the surface of the substrate depends on the functionality of the groups at the substrate surface. If the surface is amine functionalized, an amide bond is made (see example below). [0367]
    Figure US20020155491A1-20021024-C00007
  • If the surface is hydroxy functionalized, a phosphate bond is made (see example below): [0368]
    Figure US20020155491A1-20021024-C00008
  • In both cases, the thymidine example is illustrated, but any one of the four phosphoramidite activated nucleosides can be used in the first step. [0369]
  • Photolysis of the photolabile group NV or NVOC on the 5′ positions of the nucleosides is carried out at ˜362 nm with an intensity of 14 mW/cm[0370] 2 for 10 minutes with the substrate side (side containing the photolabile group) immersed in dioxane. After the coupling of the next nucleoside is complete, the photolysis is repeated followed by another coupling until the desired oligomer is obtained.
  • One of the most common 3′-O-protecting groups is the ester, in particular the acetate: [0371]
    Figure US20020155491A1-20021024-C00009
  • The groups can be removed by mild base treatment 0.1N NaOH/MeOH or K[0372] 2CO3/H2O/MeOH.
  • Another group used most often is the silyl ether: [0373]
    Figure US20020155491A1-20021024-C00010
  • These groups can be removed by neutral conditions using 1 M tetra-n-butylammonium fluoride in THF or under acid conditions. [0374]
  • With respect to photodeprotection, the nitroveratryl group could also be used to protect the 3′-position. [0375]
    Figure US20020155491A1-20021024-C00011
  • Here, light (photolysis) would be used to remove these protecting groups. [0376]
  • A variety of ethers can also be used in the protection of the 3-O-position: [0377]
    Figure US20020155491A1-20021024-C00012
  • Removal of these groups usually involves acid or catalytic methods. [0378]
  • Note that corresponding linkages and photoblocked amino acids are described in detail in Ser. No. 07/624,120, now abandoned, which is hereby incorporated herein by reference. [0379]
  • Although the specificity of interactions at particular locations will usually be homogeneous due to a homogeneous polymer being synthesized at each defined location, for certain purposes, it may be useful to have mixed polymers with a commensurate mixed collection of interactions occurring at specific defined locations, or degeneracy reducing analogues, which have been discussed above and show broad specificity in binding. Then, a positive interaction signal may result from any of a number of sequences contained therein. [0380]
  • As an alternative method of generating a matrix pattern on a substrate, preformed polymers may be individually attached at particular sites on the substrate. This may be performed by individually attaching reagents one at a time to specific positions on the matrix, a process which may be automated. See, e.g., Ser. No. 07/435,316, now abandoned, and Barrett et al. (1993) U.S. Pat. No. 5,252,743. Another way of generating a positionally defined matrix pattern on a substrate is to have individually specific reagents which interact with each specific position on the substrate. For example, oligonucleotides may be synthesized at defined locations on the substrate. Then the substrate would have on its surface a plurality of regions having homogeneous oligonucleotides attached at each position. [0381]
  • In particular, at least four different substrate preparation procedures are available for treating a substrate surface. They are the standard VLSIPS™ Technology method, polymeric substrates, Durapore™, and synthetic beads or fibers. The treatment labeled “standard VLSIPS™ Technology” method is described in Ser. No. 07/624,120, now abandoned, and involves applying amino-propyltriethoxysilane to a glass surface. [0382]
  • The polymeric substrate approach involves either of two ways of generating a polymeric substrate. The first uses a high concentration of aminopropyltriethoxysilane (2-20%) in an aqueous ethanol solution (95%). This allows the silane compound to polymerize both in solution and on the substrate surface, which provides a high density of amines on the surface of the glass. This density is contrasted with the standard VLSIPS method. This polymeric method allows for the deposition on the substrate surface of a monolayer due to the anhydrous method used with the aforementioned silane. [0383]
  • The second polymeric method involves either the coating or covalent binding of an appropriate acrylic acid polymer onto the substrate surface. In particular, e.g., in DNA synthesis, a monomer such as a hydroxypropylacrylate is used to generate a high density of hydroxyl groups on the substrate surface, allowing for the formation of phosphate bonds. An example of such a compound is shown: [0384]
    Figure US20020155491A1-20021024-C00013
  • The method using a Durapore™ membrane (Millipore) consists of a polyvinylidine difluoride coating with crosslinked polyhydroxylpropyl acrylate [PVDF-HPA]: [0385]
    Figure US20020155491A1-20021024-C00014
  • Here the building up of, e.g., a DNA oligomer, can be started immediately since phosphate bonds to the surface can be accomplished in the first step with no need for modification. A nucleotide dimer (5′-C-T-3′) has been successfully made on this substrate. [0386]
  • The fourth method utilizes synthetic beads or fibers. This would use another substrate, such as a teflon copolymer graft bead or fiber, which is covalently coated with an organic layer (hydrophilic) terminating in hydroxyl sites (commercially available from Molecular Biosystems, Inc.) This would offer the same advantage as the Durapore™ membrane, allowing for immediate phosphate linkages, but would give additional contour by the 3-dimensional growth of oligomers. [0387]
  • A matrix pattern of new reagents may be targeted to each specific oligonucleotide position by attaching a complementary oligonucleotide to which the substrate bound form is complementary. For instance, a number of regions may have homogeneous oligonucleotides synthesized at various locations. Oligonucleotide sequences complementary to each of these can be individually generated and linked to a particular specific reagents. Often these specific reagents will be antibodies. As each of these is specific for finding its complementary oligonucleotide, each of the specific reagents will bind through the oligonucleotide to the appropriate matrix position. A single step having a combination of different specific reagents being attached specifically to a particular oligonucleotide will thereby bind to its complement at the defined matrix position. The oligonucleotides will typically then be covalently attached, using, e.g., an acridine dye, for photocrosslinking. Psoralen is a commonly used acridine dye for photocrosslinking purposes, see, e.g., Song et al. (1979) [0388] Photochem. Photobiol. 29:1177-1197; Cimino et al. (1985) Ann. Rev. Biochem. 54:1151-1193; Parsons (1980) Photochem. Photobiol. 32:813-821; and Dattagupta et al. (1985) U.S. Pat. No. 4,542,102, and (1987) U.S. Pat. No. 4,713,326; each of which is hereby incorporated herein by reference. This method allows a single attachment manipulation to attach all of the specific reagents to the matrix at defined positions and results in the specific reagents being homogeneously located at defined positions. In many embodiments, the specific reagents will be antibodies.
  • In an alternative embodiment, antibody molecules may be used to specifically direct binding to defined positions on a substrate. The VLSIPS technology may be used to generate specific epitopes at each position on the substrate. Antibody molecules having specificity of interaction may be used to attach oligonucleotides, thereby avoiding the interference of internal polynucleotide sequences from binding to the substrate complementary oligonucleotides. In fact, the specificity of interaction for positional targeting may be achieved by use of nucleotide analogues which do not interact with the natural nucleotides. For example, other synthetic nucleotides have been made which undergo base pairing, thereby providing the specificity of targeting, but the synthetic nucleotides also do not interact with the natural biological nucleotides. Thus, synthetic oligonucleotides would be useful for attachment to biological nucleotides and specific targeting. Moreover, the VLSIPS synthetic processes would be useful in generating the VLSIPS substrate, and standard oligonucleotide synthesis could be applied, with minor modifications, to produce the complementary sequences which would be attached to other specific reagents. [0389]
  • D. Surface Immobilization [0390]
  • 1. Caged Biotin [0391]
  • An alternative method of attaching reagents in a positionally defined matrix pattern is to use a caged biotin system. See Barrett et al. (1993) U.S. Pat. No. 5,252,743, which is hereby incorporated herein by reference, for additional details on the chemistry and application of caged biotin embodiments. In short, the caged biotin has a photosensitive blocking moiety which prevents the combination of avidin to biotin. At positions where the photo-lithographic process has removed the blocking group, high affinity biotin sites are generated. Thus, by a sequential series of photolithographic deblocking steps interspersed with exposure of those regions to appropriate biotin containing reagents, only those locations where the deblocking takes place will form an avidin-biotin interaction. Because the avidin-biotin binding is very tight, this will usually be virtually irreversible binding. [0392]
  • 2. Crosslinked Interactions [0393]
  • The surface immobilization may also take place by photo crosslinking of defined oligonucleotides linked to specific reagents. After hybridization of the complementary oligonucleotides, the oligonucleotides may be crosslinked by a reagent by psoralen or another similar type of acridine dye. Other useful cross linking reagents are described in Dattagupta et al. (1985) U.S. Pat. No. 4,542,102, and (1987) U.S. Pat. No. 4,713,326. [0394]
  • In another embodiment, colony or phage plaque transfer of biological polymers may be transferred directly onto a silicon substrate. For example, a colony plate may be transferred onto a substrate having a generic oligonucleotide sequence which hybridizes to another generic complementary sequence contained on all of the vectors into which inserts are cloned. This will specifically only bind those molecules which are actually contained in the vectors containing the desired complementary sequence. This immobilization allows for producing a matrix onto which a sequence specific reagent can bind, or for other purposes. In a further embodiment, a plurality of different vectors each having a specific oligonucleotide attached to the vector may be specifically attached to particular regions on a matrix having a complementary oligonucleotide attached thereto. [0395]
  • VIII. Hybridization/Specific Interaction [0396]
  • A. General [0397]
  • As discussed previously in the VLSIPS™ Technology parent applications, the VLSIPS™ technology substrates may be used for screening for specific interactions with sequence specific targets or probes. [0398]
  • In addition, the availability of substrates having the entire repertoire of possible sequences of a defined length opens up the possibility of sequencing by hybridization. This sequence may be de novo determination of an unknown sequence, particularly of nucleic acid, verification of a sequence determined by another method, or an investigation of changes in a previously sequenced gene, locating and identifying specific changes. For example, often Maxam and Gilbert sequencing techniques are applied to sequences which have been determined by Sanger and Coulson. Each of those sequencing technologies have problems with resolving particular types of sequences. Sequencing by hybridization may serve as a third and independent method for verifying other sequencing techniques. See, e.g., (1988) [0399] Science 242:1245.
  • In addition, the ability to provide a large repertoire of particular sequences allows use of short subsequences and hybridization as a means to fingerprint a sample. This may be used in a nucleic acid, as well as other polymer embodiments. For example, fingerprinting to a high degree of specificity of sequence matching may be used for identifying highly similar samples, e.g., those exhibiting high homology to the selected probes. This may provide a means for determining classifications of particular sequences. This should allow determination of whether particular genomes of bacteria, phage, or even higher cells might be related to one another. [0400]
  • In addition, fingerprinting may be used to identify an individual source of biological sample. See, e.g., Lander, E. (1989) [0401] Nature, 339:501-505, and references therein. For example, a DNA fingerprint may be used to determine whether a genetic sample arose from another individual. This would be particularly useful in various sorts of forensic tests to determine, e.g., paternity or sources of blood samples. Significant detail on the particulars of genetic fingerprinting for identification purposes are described in, e.g., Morris et al. (1989) “Biostatistical evolution of evidence from continuous allele frequency distribution DNA probes in reference to disputed paternity of identity,” J. Forensic Science 34:1311-1317; and Neufeld et al. (1990) Scientific American 262:46-53; each of which is hereby incorporated herein by reference.
  • In another embodiment, a fingerprinting-like procedure may be used for classifying cell types by analyzing a pattern of specific nucleic acids present in the cell. A series of antibodies may be used to identify cell markers, e.g., proteins, usually on the cell surface, but intracellular markers may also be used. Antigens which are extracellularly expressed are preferred so cell lysis is unnecessary in the screening, but intracellular markers may also be useful. The markers will usually be proteins, but may be nucleic acids, lipids, metabolites, carbohydrates, or other cellular components. See, e.g., Winkelgren, I. (1990) [0402] Science News 136:234-237, which indicates extracellular DNA may be common, and suggesting that such might be characteristic of cell types, stage, or physiology. This may also be useful in defining the temporal stage of development of cells, e.g., stem cells or other cells which undergo temporal changes in development. For example, the stage of a cell, or group of cells, may be tested or defined by isolating a sample of MRNA from the population and testing to see what sequences are present in messenger populations. Direct samples, or amplified samples, may be used. Where particular MRNA or other nucleic acid sequences may be characteristic of or shown to be characteristic of particular developmental stages, physiological states, or other conditions, this fingerprinting method may define them. Similar sorts of fingerprinting may be used for determining T-cell classes or perhaps even to generate classification schemes for such proteins as major histocompatibility complex antigens. Thus, the ability to make these substrates allows both the generation of reagents which will be used for defining subclasses or classes of cells or other biological materials, but also provides the mechanisms for selecting those cells which may be found in defined population groups.
  • In addition to cell classification defined by such a combination of properties, typically expression of extracellular antigens, the present invention also provides the means for isolating homogeneous population of cells. Once the antigenic determinants which define a cell class have been identified, these antigens may be used in a sequential selection process to isolate only those cells which exhibit the combination of defining structural properties. [0403]
  • The present invention may also be used for mapping sequences within a larger segment. This may be performed by at least two methods, particularly in reference to nucleic acids. Often, enormous segments of DNA are subcloned into a large plurality of subsequences. Ordering these subsequences may be important in determining the overlaps of sequences upon nucleotide determinations. Mapping may be performed by immobilizing particularly large segments onto a matrix using the VLSIPS™ Technology. Alternatively, sequences may be ordered by virtue of subsequences shared by overlapping segments. See, e.g., Craig et al. (1990) [0404] Nuc. Acids Res. 18:2653-2660; Michiels et al. (1987) CABIOS 3:203-210; and Olson et al. (1986) Proc. Natl. Acad. Sci. USA 83:7826-7830.
  • B. Important Parameters [0405]
  • The extent of specific interaction between reagents immobilized to the VLSIPS™ Technology substrate and another sequence specific reagent may be modified by the conditions of the interaction. Sequencing embodiments typically require high fidelity hybridization and the ability to discriminate perfect matching from imperfect matching. Fingerprinting and mapping embodiments may be performed using less stringent conditions, depending upon the circumstances. [0406]
  • For example, the specificity of antibody/antigen interaction may depend upon such parameters as pH, salt concentration, ionic composition, solvent composition, detergent composition and concentration, and chaotropic agent concentration. See, e.g., Harlow and Lane (1988) [0407] Antibodies: A Laboratory Manual, Cold Spring Harbor Press, New York. By careful control of these parameters, the affinity of binding may be mapped across different sequences.
  • In a nucleic acid hybridization embodiment, the specificity and kinetics of hybridization have been described in detail by, e.g., Wetmur and Davidson (1968) [0408] J. Mol. Biol., 31:349-370, Britten and Kohne (1968) Science 161:529-530, and Kanehisa, (1984) Nuc. Acids Res. 12:203-213, each of which is hereby incorporated herein by reference. Parameters which are well known to affect specificity and kinetics of reaction include salt conditions, ionic composition of the solvent, hybridization temperature, length of oligonucleotide matching sequences, guanine and cytosine (GC) content, presence of hybridization accelerators, pH, specific bases found in the matching sequences, solvent conditions, and addition of organic solvents.
  • In particular, the salt conditions required for driving highly mismatched sequences to completion typically include a high salt concentration. The typical salt used is sodium chloride (NaCl), however, other ionic salts may be utilized, e.g., KCl. Depending on the desired stringency hybridization, the salt concentration will often be less than about 3 molar, more often less than 2.5 molar, usually less than about 2 molar, and more usually less than about 1.5 molar. For applications directed towards higher stringency matching, the salt concentrations would typically be lower. Ordinary high stringency conditions will utilize salt concentration of less than about 1 molar, more often less then about 750 millimolar, usually less than about 500 millimolar, and may be as low as about 250 or 150 millimolar. [0409]
  • The kinetics of hybridization and the stringency of hybridization both depend upon the temperature at which the hybridization is performed and the temperature at which the washing steps are performed. Temperatures at which steps for low stringency hybridization are desired would typically be lower temperatures, e.g., ordinarily at least about 15° C., more ordinarily at least about 20° C., usually at least about 25° C., and more usually at least about 30° C. For those applications requiring high stringency hybridization, or fidelity of hybridization and sequence matching, temperatures at which hybridization and washing steps are performed would typically be high. For example, temperatures in excess of about 35° C. would often be used, more often in excess of about 40° C., usually at least about 45° C., and occasionally even temperatures as high as about 50° C. or 60° C. or more. Of course, the hybridization of oligonucleotides may be disrupted by even higher temperatures. Thus, for stripping of targets from substrates, as discussed below, temperatures as high as 80° C., or even higher may be used. [0410]
  • The base composition of the specific oligonucleotides involved in hybridization affects the temperature of melting, and the stability of hybridization as discussed in the above references. However, the bias of GC rich sequences to hybridize faster and retain stability at higher temperatures can be compensated for by the inclusion in the hybridization incubation or wash steps of various buffers. Sample buffers which accomplish this result include the triethly-and trimethyl ammonium buffers. See, e.g., Wood et al. (1987) [0411] Proc. Natl. Acad. Sci. USA, 82:1585-1588, and Khrapko, K. et al. (1989) FEBS Letters 256:118-122.
  • The rate of hybridization can also be affected by the inclusion of particular hybridization accelerators. These hybridization accelerators include the volume exclusion agents characterized by dextran sulfate, or polyethylene glycol (PEG). Dextran sulfate is typically included at a concentration of between 1% and 40% by weight. The actual concentration selected depends upon the application, but typically a faster hybridization is desired in which the concentration is optimized for the system in question. Dextran sulfate is often included at a concentration of between 0.5% and 2% by weight or dextran sulfate at a concentration between about 0.5% and 5%. Alternatively, proteins which accelerate hybridization may be added, e.g., the recA protein found in [0412] E. coli or other homologous proteins.
  • With respect to those embodiments where specific reagents are not oligonucleotides, the conditions of specific interaction would depend on the affinity of binding between the specific reagent and its target. Typically parameters which would be of particular importance would be pH, salt concentration anion and cation compositions, buffer concentration, organic solvent inclusion, detergent concentration, and inclusion of such reagents such as chaotropic agents. In particular, the affinity of binding may be tested over a variety of conditions by multiple washes and repeat scans or by using reagents with differences in binding affinity to determine which reagents bind or do not bind under the selected binding and washing conditions. The spectrum of binding affinities may provide an additional dimension of information which may be very useful in identification purposes and mapping. [0413]
  • Of course, the specific hybridization conditions will be selected to correspond to a discriminatory condition which provides a positive signal where desired but fails to show a positive signal at affinities where interaction is not desired. This may be determined by a number of titration steps or with a number of controls which will be run during the hybridization and/or washing steps to determine at what point the hybridization conditions have reached the stage of desired specificity. [0414]
  • IX. Detection Methods [0415]
  • Methods for detection depend upon the label selected. The criteria for selecting an appropriate label are discussed below, however, a fluorescent label is preferred because of its extreme sensitivity and simplicity. Standard labeling procedures are used to determine the positions where interactions between a sequence and a reagent take place. For example, if a target sequence is labeled and exposed to a matrix of different probes, only those locations where probes do interact with the target will exhibit any signal. Alternatively, other methods may be used to scan the matrix to determine where interaction takes place. Of course, the spectrum of interactions may be determined in a temporal manner by repeated scans of interactions which occur at each of a multiplicity of conditions. However, instead of testing each individual interaction separately, a multiplicity of sequence interactions may be simultaneously determined on a matrix. [0416]
  • A. Labeling Technicues [0417]
  • The target polynucleotide may be labeled by any of a number of convenient detectable markers. A fluorescent label is preferred because it provides a very strong signal with low background. It is also optically detectable at high resolution and sensitivity through a quick scanning procedure. Other potential labeling moieties include, radioisotopes, chemiluminescent compounds, labeled binding proteins, heavy metal atoms, spectroscopic markers, magnetic labels, and linked enzymes. [0418]
  • Another method for labeling may bypass any label of the target sequence. The target may be exposed to the probes, and a double strand hybrid is formed at those positions only. Addition of a double strand specific reagent will detect where hybridization takes place. An intercalative dye such as ethidium bromide may be used as long as the probes themselves do not fold back on themselves to a significant extent forming hairpin loops. See, e.g., Sheldon et al. (1986) U.S. Pat. No. 4,582,789. However, the length of the hairpin loops in short oligonucleotide probes would typically be insufficient to form a stable duplex. [0419]
  • In another embodiment, different targets may be simultaneously sequenced where each target has a different label. For instance, one target could have a green fluorescent label and a second target could have a red fluorescent label. The scanning step will distinguish sites of binding of the red label from those binding the green fluorescent label. Each sequence can be analyzed independently from one another. [0420]
  • Suitable chromogens will include molecules and compounds which absorb light in a distinctive range of wavelengths so that a color may be observed, or emit light when irradiated with radiation of a particular wave length or wave length range, e.g., fluorescers. Biliproteins, e.g., phycoerythrin. may also serve as labels. [0421]
  • A wide variety of suitable dyes are available, being primarily chosen to provide an intense color with minimal absorption by their surroundings. Illustrative dye types include quinoline dyes, triarylmethane dyes, acridine dyes, alizarine dyes, phthaleins, insect dyes, azo dyes, anthraquinoid dyes, cyanine dyes, phenazathionium dyes, and phenazoxonium dyes. [0422]
  • A wide variety of fluorescers may be employed either by themselves or in conjunction with quencher molecules. Fluorescers of interest fall into a variety of categories having certain primary functionalities. These primary functionalities include 1- and 2-aminonaphthalene, p,p′-diaminostilbenes, pyrenes, quaternary phenanthridine salts, 9-aminoacridines, p,p′-diaminobenzophenone imines, anthracenes, oxacarbocyanine, merocyanine, 3-aminoequilenin, perylene, bis-benzoxazole, bis-p-oxazolyl benzene, 1,2-benzophenazin, retinol, bis-3-aminopyridinium salts, hellebrigenin, tetracycline, sterophenol, benzimidzaolylphenylamine, 2-oxo-3-chromen, indole, xanthen, 7-hydroxycoumarin, phenoxazine, salicylate, strophanthidin, porphyrins, triarylmethanes and flavin. Individual fluorescent compounds which have functionalities for linking or which can be modified to incorporate such functionalities include, e.g., dansyl chloride; fluoresceins such as 3,6-dihydroxy-9-phenylxanthhydrol; rhodamineisothiocyanate; N-phenyl 1-amino-8-sulfonatonaphthalene; N-phenyl 2-amino-6-sulfonatonaphthalene; 4-acetamido-4-isothiocyanato-stilbene-2,2′-disulfonic acid; pyrene-3-sulfonic acid; 2-toluidinonaphthalene-6-sulfonate; N-phenyl, N-methyl 2-aminoaphthalene-6-sulfonate; ethidium bromide; stebrine; auromine-0,2-(9′-anthroyl)palmitate; dansyl phosphatidylethanolamine; N,N′-dioctadecyl oxacarbocyanine; N,N′-dihexyl oxacarbocyanine; merocyanine, 4-(3′pyrenyl)butyrate; d-3-aminodesoxy-equilenin; 12-(9′-anthroyl)stearate; 2-methylanthracene; 9-vinylanthracene; 2,2′-(vinylene-p-phenylene)bisbenzoxazole; p-bis[2-(4-methyl-5-phenyl-oxazolyl)]benzene; 6-dimethylamino-1,2-benzophenazin; retinol; bis(3′-aminopyridinium) 1,10-decandiyl diiodide; sulfonaphthylhydrazone of hellibrienin; chlorotetracycline; N-(7-dimethylamino-4-methyl-2-oxo-3-chromenyl)maleimide; N-[p-(2-benzimidazolyl)-phenyl]maleimide; N-(4-fluoranthyl)maleimide; bis(homovanillic acid); resazarin; 4-chloro-7-nitro-2,1,3-benzooxadiazole; merocyanine 540; resorufin; rose bengal; and 2,4-diphenyl-3(2H)-furanone. [0423]
  • Desirably, fluorescers should absorb light above about 300 nm, preferably about 350 nm, and more preferably above about 400 nm, usually emitting at wavelengths greater than about 10 nm higher than the wavelength of the light absorbed. It should be noted that the absorption and emission characteristics of the bound dye may differ from the unbound dye. Therefore, when referring to the various wavelength ranges and characteristics of the dyes, it is intended to indicate the dyes as employed and not the dye which is unconjugated and characterized in an arbitrary solvent. [0424]
  • Fluorescers are generally preferred because by irradiating a fluorescer with light, one can obtain a plurality of emissions. Thus, a single label can provide for a plurality of measurable events. [0425]
  • Detectable signal may also be provided by chemiluminescent and bioluminescent sources. Chemiluminescent sources include a compound which becomes electronically excited by a chemical reaction and may then emit light which serves as the detectible signal or donates energy to a fluorescent acceptor. A diverse number of families of compounds have been found to provide chemiluminescence under a variety of conditions. One family of compounds is 2,3-dihydro-1,-4-phthalazinedione. The most popular compound is luminol, which is the 5-amino compound. Other members of the family include the 5-amino-6,7,8-trimethoxy- and the dimethylamino[ca]benz analog. These compounds can be made to luminesce with alkaline hydrogen peroxide or calcium hypochlorite and base. Another family of compounds is the 2,4,5-triphenylimidazoles, with lophine as the common name for the parent product. Chemiluminescent analogs include para-dimethylamino and -methoxy substituents. Chemiluminescence may also be obtained with oxalates, usually oxalyl active esters, e.g., p-nitrophenyl and a peroxide, e.g., hydrogen peroxide, under basic conditions. Alternatively, luciferins may be used in conjunction with luciferase or lucigenins to provide bioluminescence. [0426]
  • Spin labels are provided by reporter molecules with an unpaired electron spin which can be detected by electron spin resonance (ESR) spectroscopy. Exemplary spin labels include organic free radicals, transitional metal complexes, particularly vanadium, copper, iron, and manganese, and the like. Exemplary spin labels include nitroxide free radicals. [0427]
  • B. Scanning System [0428]
  • With the automated detection apparatus, the correlation of specific positional labeling is converted to the presence on the target of sequences for which the reagents have specificity of interaction. Thus, the positional information is directly converted to a database indicating what sequence interactions have occurred. For example, in a nucleic acid hybridization application, the sequences which have interacted between the substrate matrix and the target molecule can be directly listed from the positional information. The detection system used is described in Pirrung et al. (1992) U.S. Pat. No. 5,143,854; and Ser. No. 07/624,120, now abandoned. Although the detection described therein is a fluorescence detector, the detector may be replaced by a spectroscopic or other detector. The scanning system may make use of a moving detector relative to a fixed substrate, a fixed detector with a moving substrate, or a combination. Alternatively, mirrors or other apparatus can be used to transfer the signal directly to the detector. See, e.g, Ser. No. 07/624,120, now abandoned, which is hereby incorporated herein by reference. [0429]
  • The detection method will typically also incorporate some signal processing to determine whether the signal at a particular matrix position is a true positive or may be a spurious signal. For example, a signal from a region which has actual positive signal may tend to spread over and provide a positive signal in an adjacent region which actually should not have one. This may occur, e.g., where the scanning system is not properly discriminating with sufficiently high resolution in its pixel density to separate the two regions. Thus, the signal over the spatial region may be evaluated pixel by pixel to determine the locations and the actual extent of positive signal. A true positive signal should, in theory, show a uniform signal at each pixel location. Thus, processing by plotting number of pixels with actual signal intensity should have a clearly uniform signal intensity. Regions where the signal intensities show a fairly wide dispersion, may be particularly suspect and the scanning system may be programmed to more carefully scan those positions. [0430]
  • In another embodiment, as the sequence of a target is determined at a particular location, the overlap for the sequence would necessarily have a known sequence. Thus, the system can compare the possibilities for the next adjacent position and look at these in comparison with each other. Typically, only one of the possible adjacent sequences should give a positive signal and the system might be programmed to compare each of these possibilities and select that one which gives a strong positive. In this way, the system can also simultaneously provide some means of measuring the reliability of the determination by indicating what the average signal to background ratio actually is. [0431]
  • More sophisticated signal processing techniques can be applied to the initial determination of whether a positive signal exists or not. See, e.g., Ser. No. 07/624,120, now abandoned. [0432]
  • From a listing of those sequences which interact, data analysis may be performed on a series of sequences. For example, in a nucleic acid sequence application, each of the sequences may be analyzed for their overlap regions and the original target sequence may be reconstructed from the collection of specific subsequences obtained therein. Other sorts of analyses for different applications may also be performed, and because the scanning system directly interfaces with a computer the information need not be transferred manually. This provides for the ability to handle large amounts of data with very little human intervention. This, of course, provides significant advantages over manual manipulations. Increased throughput and reproducibility is thereby provided by the automation of a vast majority of steps in any of these applications. [0433]
  • XI. Data Analysis [0434]
  • A. General [0435]
  • Data analysis will typically involve aligning the proper sequences with their overlaps to determine the target sequence. Although the target “sequence” may not specifically correspond to any specific molecule, especially where the target sequence is broken and fragmented in the sequencing process, the sequence corresponds to a contiguous sequence of the subfragments. [0436]
  • The data analysis can be performed by a computer using an appropriate program. See, e.g., Drmanac, R. et al. (1989) [0437] Genomics 4:114-128; and a commercially available analysis program available from the Genetic Engineering Center, P.O. Box 794, 11000 Belgrade, Yugoslavia. Although the specific manipulations necessary to reassemble the target sequence from fragments may take many forms, one embodiment uses a sorting program to sort all of the subsequences using a defined hierarchy. The hierarchy need not necessarily correspond to any physical hierarchy, but provides a means to determine, in order, which subfragments have actually been found in the target sequence. In this manner, overlaps can be checked and found directly rather than having to search throughout the entire set after each selection process. For example, where the oligonucleotide probes are 10-mers, the first 9 positions can be sorted. A particular subsequence can be selected as in the examples, to determine where the process starts. As analogous to the theoretical example provided above, the sorting procedure provides the ability to immediately find the position of the subsequence which contains the first 9 positions and can compare whether there exists more than 1 subsequence during the first 9 positions. In fact, the computer can easily generate all of the possible target sequences which contain given combination of subsequences. Typically there will be only one, but in various situations, there will be more.
  • An exemplary flow chart for a sequencing program is provided in FIG. 1. In general terms, the program provides for automated scanning of the substrate to determine the positions of probe and target interaction. Simple processing of the intensity of the signal may be incorporated to filter out clearly spurious signals. The positions with positive interaction are correlated with the sequence specificity of specific matrix positions, to generate the set of matching subsequences. This information is further correlated with other target sequence information, e.g., restriction fragment analysis. The sequences are then aligned using overlap data, thereby leading to possible corresponding target sequences which will, optimally, correspond to a single target sequence. [0438]
  • B. Hardware [0439]
  • A variety of computer systems may be used to run a sequencing program. The program may be written to provide both the detecting and scanning steps together and will typically be dedicated to a particular scanning apparatus. However, the components and functional steps may be separated and the scanning system may provide an output, e.g., through tape or an electronic connection into a separate computer which separately runs the sequencing analysis program. The computer may be any of a number of machines provided by standard computer manufacturers, e.g., IBM compatible machines, Apple™ machines, VAX machines, and others, which may often use a UNIX™ operating system. Of course, the hardware used to run the analysis program will typically determine what programming language would be used. [0440]
  • C. Software [0441]
  • Software would be easily developed by a person of ordinary skill in the programming art, following the flow chart provided, or based upon the input provided and the desired result. [0442]
  • Of course, an exemplary embodiment is a polynucleotide sequence system. However, the theoretical and mathematical manipulations necessary for data analysis of other linear molecules, such as polypeptides, carbohydrates, and various other polymers are conceptually similar. Simple branching polymers will usually also be sequencable using similar technology. However, where there is branching, it may be desired that additional recognition reagents be used to determine the nature and location of branches. This can easily be provided by use of appropriate specific reagents which would be generated by methods similar to those used to produce specific reagents for linear polymers. [0443]
  • XII. Substrate Reuse [0444]
  • Where a substrate is made with specific reagents that are relatively insensitive to the handling and processing steps involved in a single cycle of use, the substrate may often be reused. The target molecules are usually stripped off of the solid phase specific recognition molecules. Of course, it is preferred that the manipulations and conditions be selected as to be mild and to not affect the substrate. For example, if a substrate is acid labile, a neutral pH would be preferred in all handling steps. similar sensitivities would be carefully respected where recycling is desired. [0445]
  • A. Removal of Label [0446]
  • Typically for a recycling, the previously attached specific interaction would be disrupted and removed. This will typically involve exposing the substrate to conditions under which the interaction between probe and target is disrupted. Alternatively, it may be exposed to conditions where the target is destroyed. For example, where the probes are oligonucleotides and the target is a polynucleotide, a heating and low salt wash will often be sufficient to disrupt the interactions. Additional reagents may be added such as detergents, and organic or inorganic solvents which disrupt the interaction between the specific reagents and target. In an embodiment where the specific reagents are antibodies, the substrate may be exposed to a gentle detergent which will denature the specific binding between the antibody and its target. The conditions are selected to avoid severe disruption or destruction of the structure of the antibody and to maintain the specificity of the antibody binding site. Conditions with specific pH, detergent concentration, salt concentration, ionic concentration, and other parameters may be selected which disrupt the specific interactions. [0447]
  • B. Storage and Preservation [0448]
  • As indicated above, the matrix will typically be maintained under conditions where the matrix itself and the linkages and specific reagents are preserved. Various specific preservatives may be added which prevent degradation. For example, if the reagents are acid or base labile, a neutral pH buffer will typically be added. It is also desired to avoid destruction of the matrix by growth of organisms which may destroy organic reagents attached thereto. For this reason, a preservative such as cyanide or azide may be added. However, the chemical preservative should also be selected to preserve the chemical nature of the linkages and other components of the substrate. Typically, a detergent may also be included. [0449]
  • C. Processes to Avoid Degradation of Oliaomers [0450]
  • In particular, a substrate comprising a large number of oligomers will be treated in a fashion which is known to maintain the quality and integrity of oligonucleotides. These include storing the substrate in a carefully controlled environment under conditions of lower temperature, cation depletion (EDTA and EGTA), sterile conditions, and inert argon or nitrogen atmosphere. [0451]
  • XIII. Integrated Sequencing Strategy [0452]
  • A. Initial Mapping Strategy [0453]
  • As indicated above, although the VLSIPS™ technology may be applied to sequencing embodiments, it is often useful to integrate other concepts to simplify the sequencing. For example, nucleic acids may be easily sequenced by careful selection of the vectors and hosts used for amplifying and generating the specific target sequences. For example, it may be desired to use specific vectors which have been designed to interact most efficiently with the VLSIPS substrate. This is also important in fingerprinting and mapping strategies. For example, vectors may be carefully selected having particular complementary sequences which are designed to attach to a genetic or specific oligomer on the substrate. This is also applicable to situations where it is desired to target particular sequences to specific locations on the matrix. [0454]
  • In one embodiment, unnatural oligomers may be used to target natural probes to specific locations on the VLSIPS substrate. In addition, particular probes may be generated for the mapping embodiment which are designed to have specific combinations of characteristics. For example, the construction of a mapping substrate may depend upon use of another automated apparatus which takes clones isolated from a chromosome walk and attaches them individually or in bulk to the VLSIPS substrate. [0455]
  • In another embodiment, a variety of specific vectors having known and particular “targeting” sequences adjacent to the cloning sites may be individually used to clone a selected probe, and the isolated probe will then be targetable to a site on the VLSIPS substrate with a sequence complementary to the “target” sequence. [0456]
  • B. selection of Smaller Clones [0457]
  • In the fingerprinting and mapping embodiments, the selection of probes may be very important. Significant mathematical analysis may be applied to determine which specific sequences should be used as those probes. Of course, for fingerprinting use, these sequences would be most desired that show significant heterogeneity across the human population. Selection of the specific sequences which would most favorably be utilized will tend to be single copy sequences within the genome. [0458]
  • Various hybridization selection procedures may be applied to select sequences which tend not to be repeated within a genome, and thus would tend to be conserved across individuals. For example, hybridization selections may be made for non-repetitive and single copy sequences. See, e.g., Britten and Kohne (1968) “Repeated Sequences in DNA,” [0459] Science 161:529-540. On the other hand, it may be desired under certain circumstances to use repeated sequences. For example, where a fingerprint may be used to identify or distinguish different species, or where repetitive sequences may be diagnostic of specific species, repetitive sequences may be desired for inclusion in the fingerprinting probes. In either case, the sequencing capability will greatly assist in the selection of appropriate sequences to be used as probes.
  • Also as indicated above, various means for constructing an appropriate substrate may involve either mechanical or automated procedures. The standard VLSIPS automated procedure involves synthesizing oligonucleotides or short polymers directly on the substrate. In various other embodiments, it is possible to attach separately synthesized reagents onto the matrix in an ordered array. Other circumstances may lend themselves to transfer a pattern from a petri plate onto a solid substrate. Also, there are methods for site specifically directing collections of reagents to specific locations using unnatural nucleotides or equivalent sorts of targeting molecules. [0460]
  • While a brute force manual transfer process may be utilized sequentially for attaching various samples to successive positions, instrumentation for automating such procedures may also be devised. The automated system for performing such would preferably be relatively easily designed and conceptually easily understood. [0461]
  • XIV. Commercial Applications [0462]
  • A. Sequencing [0463]
  • As indicated above, sequencing may be performed either de novo or as a verification of another sequencing method. The present hybridization technology provides the ability to sequence nucleic acids and polynucleotides de novo, or as a means to verify either the maxam and Gilbert chemical sequencing technique or Sanger and Coulson dideoxy- sequencing techniques. The hybridization method is useful to verify sequencing determined by any other sequencing technique and to closely compare two similar sequences, e.g., to identify and locate sequence differences. [0464]
  • Besides polynucleotide sequencing, the present invention also provides means for sequencing other polymers. This includes polypeptides, carbohydrates, synthetic organic polymers, and other polymers. Again, the sequencing may be either verification or de novo. [0465]
  • Of course, sequencing can be very important in many different sorts of environments. For example, it will be useful in determining the genetic sequence of particular markers in various individuals. In addition, polymers may be used as markers or for information containing molecules to encode information. For example, a short polynucleotide sequence may be included in large bulk production samples indicating the manufacturer, date, and location of manufacture of a product. For example, various drugs may be encoded with this information with a small number of molecules in a batch. For example, a pill may have somewhere from 10 to 100 to 1,000 or more very short and small molecules encoding this information. When necessary, this information may be decoded from a sample of the material using a polymerase chain reaction (PCR) or other amplification method. This encoding system may be used to provide the origin of large bulky samples without significantly affecting the properties of those samples. For example, chemical samples may also be encoded by this method thereby providing means for identifying the source and manufacturing details of lots. The origin of bulk hydrocarbon samples may be encoded. Production lots of organic compounds such as benzene or plastics may be encoded with a short molecule polymer. Food stuffs may also be encoded using similar marking molecules. Even toxic waste samples can be encoded determining the source or origin. In this way, proper disposal can be traced or more easily enforced. [0466]
  • Similar sorts of encoding may be provided by fingerprinting-type analysis. Whether the resolution is absolute or less so, the concept of coding information on molecules such as nucleic acids, which can be amplified and later decoded, may be a very useful and important application. [0467]
  • This technology also provides the ability to include markers for origins of biological materials. For example, a patented animal line may be transformed with a particular unnatural sequence which can be traced back to its origin. With a selection of multiple markers, the likelihood could be negligible that a combination of markers would have independently arisen from a source other than the patented or specifically protected source. This technique may provide a means for tracing the actual origin of particular biological materials. Bacteria, plants, and animals will be subject to marking by such encoding sequences. [0468]
  • B. Fingerprinting [0469]
  • As indicated above, fingerprinting technology may also be used for data encryption. Moreover, fingerprinting allows for significant identification of particular individuals. Where the fingerprinting technology is standardized, and used for identification of large numbers of people, related equipment and peripheral processing will be developed to accompany the underlying technology. For example, specific equipment may be developed for automatically taking a biological sample and generating or amplifying the information molecules within the sample to be used in fingerprinting analysis. Moreover, the fingerprinting substrate may be mass produced using particular types of automatic equipment. Synthetic equipment may produce the entire matrix simultaneously by stepwise synthetic methods as provided by the VLSIPS™ technology. The attachment of specific probes onto a substrate may also be automated, e.g., making use of the caged biotin technology. See, e.g., Barrett et al. (1993) U.S. Pat. No. 5,252,743. As indicated above, there are automated methods for actually generating the matrix and substrate with distinct sequence reagents positionally located at each of the matrix positions. Where such reagents are, e.g., unnatural amino acids, a targeting function may be utilized which does not interfere with a natural nucleotide functionality. [0470]
  • In addition, peripheral processing may be important and may be dedicated to this specific application. Thus, automated equipment for producing the substrates may be designed, or particular systems which take in a biological sample and output either a computer readout or an encoded instrument, e.g., a card or document which indicates the information and can provide that information to others. An identification having a short magnetic strip with a few million bits may be used to provide individual identification and important medical information useful in a medical emergency. [0471]
  • In fact, data banks may be set up to correlate all of this information of fingerprinting with medical information. This may allow for the determination of correlations between various medical problems and specific DNA sequences. By collating large populations of medical records with genetic information, genetic propensities and genetic susceptibilities to particular medical conditions may be developed. Moreover, with standardization of substrates, the micro encoding data may be also standardized to reproduce the information from a centralized data bank or on an encoding device carried on an individual person. On the other hand, if the fingerprinting procedure is sufficiently quick and routine, every hospital may routinely perform a fingerprinting operation and from that determine many important medical parameters for an individual. [0472]
  • In particular industries, the VLSIPS sequencing, fingerprinting, or mapping technology will be particularly appropriate. As mentioned above, agricultural livestock suppliers may be able to encode and determine whether their particular strains are being used by others. By incorporating particular markers into their genetic stocks, the markers will indicate origin of genetic material. This is applicable to seed producers, livestock producers, and other suppliers of medical or agricultural biological materials. [0473]
  • This may also be useful in identifying individual animals or plants. For example, these markers may be useful in determining whether certain fish return to their original breeding grounds, whether sea turtles always return to their original birthplaces, or to determine the migration patterns and viability of populations of particular endangered species. It would also provide means for tracking the sources of particular animal products. For example, it might be useful for determining the origins of controlled animal substances such as elephant ivory or particular bird populations whose importation or exportation is controlled. [0474]
  • As indicated above, polymers may be used to encode important information on source and batch and supplier. This is described in greater detail, e.g., “Applications of PCR to industrial problems,” (1990) in [0475] Chemical and Engineering News 68:145, which is hereby incorporated herein by reference. In fact, the synthetic method can be applied to the storage of enormous amounts of information. Small substrates may encode enormous amounts of information, and its recovery will make use of the inherent replication capacity. For example, on regions of 10 μm×10 μm, 1 cm2 has 106 regions. In theory, the entire human genome could be attached in 1000 nucleotide segments on a 3 cm2 surface. Genomes of endangered species may be stored on these substrates.
  • Fingerprinting may also be used for genetic tracing or for identifying individuals for forensic science purposes. See, e.g., Morris, J. et al. (1989) “Biostatistical Evaluation of Evidence From Continuous Allele Frequency Distribution DNA Probes in Reference to Disputed Paternity and Identity,” [0476] J. Forensic Science 34:1311-1317, and references provided therein; each of which is hereby incorporated herein by reference.
  • In addition, the high resolution fingerprinting allows the distinguishability to high resolution of particular samples. As indicated above, new cell classifications may be defined based on combinations of a large number of properties. Similar applications will be found in distinguishing different species of animals or plants. In fact, microbial identification may become dependent on characterization of the genetic content. Tumors or other cells exhibiting abnormal physiology will be detectable by use of the present invention. Also, knowing the genetic fingerprint of a microorganism may provide very useful information on how to treat an infection by such organism. [0477]
  • Modifications of the fingerprint embodiments may be used to diagnose the condition of the organism. For example, a blood sample is presently used for diagnosing any of a number of different physiological conditions. A multi-dimensional fingerprinting method ade available by the present invention could become a routine means for diagnosing an enormous number of physiological features simultaneously. This may revolutionize the practice of medicine in providing information on an enormous number of parameters together at one time. In another way, the genetic predisposition may also revolutionize the practice of medicine providing a physician with the ability to predict the likelihood of particular medical conditions arising at any particular moment. It also provides the ability to apply preventive medicine. [0478]
  • The present invention might also find application in use for screening new drugs and new reagents which may be very important in medical diagnosis or other applications. For example, a description of generating a population of monoclonal antibodies with defined specificities may be very useful for producing various drugs or diagnostic reagents. [0479]
  • Also available are kits with the reagents useful for performing sequencing, fingerprinting, and mapping procedures. The kits will have various compartments with the desired necessary reagents, e.g., substrate, labeling reagents for target samples, buffers, and other useful accompanying products. [0480]
  • C. Mapping [0481]
  • The present invention also provides the means for mapping sequences within enormous stretches of sequence. For example, nucleotide sequences may be mapped within enormous chromosome size sequence maps. For example, it would be possible to map a chromosomal location within the chromosome which contains hundreds of millions of nucleotide base pairs. In addition, the mapping and fingerprinting embodiments allow for testing of chromosomal translocations, one of the standard problems for which amniocentesis is performed. [0482]
  • Thus, the present invention provides a powerful tool and the means for performing sequencing, fingerprinting, and mapping functions on polymers. Although most easily and directly applicable to polynucleotides, polypeptides, carbohydrates, and other sorts of molecules can be advantageously utilized using the present technology. [0483]
  • The present invention will be better understood by reference to the following illustrative examples. The following examples are offered by way of illustration and not by way of limitation. [0484]
  • Experimental
  • I. Sequencing [0485]
  • A. polynucleotide [0486]
  • B. polypeptide [0487]
  • C. short peptide [0488]
  • 1. Herz antibody identification [0489]
  • II. Fingerprinting [0490]
  • A. polynucleotide fingerprint [0491]
  • B. peptide fingerprint [0492]
  • C. cell classification scheme [0493]
  • D. temporal development scheme [0494]
  • 1. developmental antigens [0495]
  • 2. developmental MRNA expression [0496]
  • E. diagnostic test [0497]
  • 1. viral identification [0498]
  • 2. bacterial identification [0499]
  • 3. other microbiological identifications [0500]
  • 4. allergy test (immobilized antigens) [0501]
  • F. individual (animal/plant) identification [0502]
  • 1. genetic [0503]
  • 2. immunological [0504]
  • G. genetic screen [0505]
  • 1. test alleles with markers [0506]
  • 2. amniocentesis [0507]
  • III. Mapping [0508]
  • A. positionally located clones (caged biotin) [0509]
  • 1. short probes, long targets [0510]
  • 2. long targets, short probes [0511]
  • B. positionally defined clones [0512]
  • IV. Conclusion [0513]
  • Relevant applications whose techniques are incorporated herein by reference are Pirrung, et al., Ser. No. 07/362,901, filed Jun. 7, 1989, now abandoned; Pirrung et al. (1992) U.S. Pat. No. 5,143,854; Barrett, et al., Ser. No. 07/435,316 filed Nov. 13, 1989, now abandoned; Barrett, et al. (1993) U.S. Pat. No. 5,252,743; and commonly assigned and simultaneously filed applications Ser. No. 07/624,120, now abandoned, and Ser. No. 07/626,730. [0514]
  • Also, additional relevant techniques are described, e.g., in Sambrook, J., et al. (1989) [0515] Molecular Cloninc: a Laboratory Manual, 2d Ed., vols 1-3, Cold Spring Harbor Press, New York; Greenstein and Winitz (1961) Chemistry of the Amino Acids, Wiley and Sons, New York; Bodzansky, M. (1988) Peptide Chemistry: a Practical Textbook, Springer-Verlag, New York; Harlow and Lane (1988) Antibodies: A Laboratory Manual, Cold Spring Harbor Press, New York; Glover, D. (ed.) (1987) DNA Cloning: A Practical Approach, vols 1-3, IRL Press, Oxford; Bishop and Rawlings (1987) Nucleic Acid and Protein Sequence Analysis: A Practical Approach, IRL Press, Oxford; Hames and Higgins (1985) Nucleic Acid Hybridisation: A Practical Approach, IRL Press, Oxford; Wu et al. (1989) Recombinant DNA Methodology, Academic Press, San Diego; Goding (1986) Monoclonal Antibodies: Principles and Practice, (2d ed.), Academic Press, San Diego; Finegold and Barron (1986) Bailey and Scott's Diagnostic Microbiology, (7th ed.), Mosby Co., St. Louis; Collins et al. (1989) Microbiological Methods, (6th ed.), Butterworth, London; Chaplin and Kennedy (1986) Carbohydrate Analysis: A Practical Approach, IRL Press, Oxford; Van Dyke (ed.) (1985) Bioluminescence and Chemiluminescence: Instruments and Applications, vol 1, CRC Press, Boca Rotan; and Ausubel et al. (ed.) (1990) Current Protocols in Molecular Biology, Greene Publishing and Wiley-Interscience, New York; each of which is hereby incorporated herein by reference.
  • The following examples are provided to illustrate the efficacy of the inventions herein. All operations were conducted at about ambient temperatures and pressures unless indicated to the contrary. [0516]
  • I. Sequencing [0517]
  • A. Polynucleotide [0518]
  • 1. HPLC of the Photolysis of 5′-O-nitroveratryl-thymidine [0519]
  • In order to determine the time for photolysis of 5′-O-nitroveratryl thymidine to thymidine a 100 μM solution of NV-Thym-OH (5′-O-nitroveratryl thymidine) in dioxane was made and ˜200 μl aliquots were irradiated (in a quartz cuvette 1 cm×2 mm) at 362.3 nm for 20 sec, 40 sec, 60 sec, 2 min, 5 min, 10 min, 15 min, and 20 min. The resulting irradiated mixtures were then analyzed by HPLC using a Varian MicroPak SP column (C[0520] 18 analytical) at a flow rate of 1 ml/min and a solvent system of 40% CH3CN and 60% water. Thymidine has a retention time of 1.2 min and NVO-Thym-OH has a retention time of 2.1 min. It was seen that after 10 min of exposure the deprotection was complete.
  • 2. Preparation and Detection of Thymidine-Cytidine Dimer (FITC) [0521]
  • The reaction is illustrated: [0522]
    Figure US20020155491A1-20021024-C00015
  • To an aminopropylated glass slide (standard VLSIPS™ Technology) was added a mixture of the following: [0523]
  • 12.2 mg of NVO-Thym-CO[0524] 2H (IX)
  • 3.4 mg of HOBT (N-hydroxybenztriazal) [0525]
  • 8.8 μl DIEA (Diisopropylethylamine) [0526]
  • 11.1 mg BOP reagent [0527]
  • 2.5 ml DMF [0528]
  • After 2 h coupling time (standard VLSIPS) the plate was washed, acetylated with acetic anhydride/pyridine, washed, dried, and photolyzed in dioxane at 362 nm at 14 mW/cm[0529] 2 for 10 min using a 500 μm checkerboard mask. The slide was then taken and treated with a mixture of the following:
  • 107 mg of FMOC-amine modified C (III) [0530]
  • 21 mg of tetrazole [0531]
  • 1 ml anhydrous CH[0532] 3CN
  • After being treated for approximately 8 min, the slide was washed off with CH[0533] 3CN, dried, and oxidized with I2/H2O/THF/lutidine for 1 min. The slide was again washed, dried, and treated for 30 min with a 20% solution of DBU in DMF. After thorough rinsing of the slide, it was next exposed to a FITC solution (1 mM fluorescein isothiocyanate [FITC] in DMF) for 50 min, then washed, dried, and examined by fluorescence microscopy. This reaction is illustrated:
    Figure US20020155491A1-20021024-C00016
  • Preparation and Detection of Thymidine-Cytidine Dimer (Biotin) [0534]
  • An aminopropyl glass slide, was soaked in a solution of ethylene oxide (20% in DMF) to generate a hydroxylated surface. The slide was added to a mixture of the following: [0535]
  • 32 mg of NVO-T-OCED (X) [0536]
  • 11 mg of tetrazole [0537]
  • 0.5 ml of anhydrous CH[0538] 3CN
  • After 8 min the plate was then rinsed with acetonitrile, then oxidized with I[0539] 2/H2O/THF/lutidine for 1 min, washed and dried. The slide was then exposed to a 1:3 mixture of acetic anhydride:pyridine for 1 h, then washed and dried. The substrate was then photolyzed in dioxane at 362 nm at 14 mW/cm2 for 10 min using a 500 μm checkerboard mask, dried, and then treated with a mixture of the following:
  • 65 mg of biotin modified C (IV) [0540]
  • 11 mg of tetrazole [0541]
  • 0.5 ml anhydrous CH[0542] 3CN
  • After 8 min the slide was washed with CH[0543] 3CN then oxidized with I2/H2O/THF/lutidine for 1 min, washed, and then dried. The slide was then soaked for 30 min in a PBS/0.05% Tween 20 buffer and the solution then shaken off. The slide was next treated with FITC-labeled streptavidin at 10 μg/ml in the same buffer system for 30 min. After this time the streptavidin-buffer system was rinsed off with fresh PBS/0.05% Tween 20 buffer and then the slide was finally agitated in distilled water for about ½ h. After drying, the slide was examined by fluorescence microscopy.
  • 4. Substrate Preparation [0544]
  • Before attachment of reactive groups it is preferred to clean the substrate which is, in a preferred embodiment, a glass substrate such as a microscope slide or cover slip. A roughened surface will be useable but a plastic or other solid substrate is also appropriate. According to one embodiment the slide is soaked in an alkaline bath consisting of, e.g., 1 liter of 95% ethanol with 120 ml of water and 120 grams of sodium hydroxide for 12 hours. The slides are washed with a buffer and under running water, allowed to air dry, and rinsed with a solution of 95% ethanol. [0545]
  • The slides are then aminated with, e.g., aminopropyltriethoxysilane for the purpose of attaching amino groups to the glass surface on linker molecules, although other omega functionalized silanes could also be used for this purpose. In one embodiment 0.1% aminopropyltriethoxysilane is utilized, although solutions with concentrations from 10[0546] −2% to 10% may be used, with about 10−3% to 2% preferred. A 0.1% mixture is prepared by adding to 100 ml of a 95% ethanol/5% water mixture, 100 microliters (μl) of aminopropyltriethoxysilane. The mixture is agitated at about ambient temperature on a rotary shaker for an appropriate amount of time, e.g., about 5 minutes. 500 μl of this mixture is then applied to the surface of one side of each cleaned slide. After 4 minutes or more, the slides are decanted of this solution and thoroughly rinsed three times or more by dipping in 100% ethanol.
  • After the slides dry, they are heated in a 110-120° C. vacuum oven for about 20 minutes, and then allowed to cure at room temperature for about 12 hours in an argon environment. The slides are then dipped into DMF (dimethylformamide) solution, followed by a thorough washing with methylene chloride. [0547]
  • 5. Linker Attachment, Blocking of Free Sites [0548]
  • The aminated surface of the slide is then exposed to about 500 μl of, for example, a 30 millimolar (mM) solution of NVOC-nucleotide-NHS (N-hydroxysuccinimide) in DMF for attachment of a NVOC-nucleotide to each of the amino groups. See, e.g., SIGMA Chemical Company for various nucleotide derivatives. The surface is washed with, for example, DMF, methylene chloride, and ethanol. [0549]
  • Any unreacted aminopropyl silane on the surface, i.e., those amino groups which have not had the NVOC-nucleotide attached, are now capped with acetyl groups (to prevent further reaction) by exposure to a 1:3 mixture of acetic anhydride in pyridine for 1 hour. Other materials which may perform this residual capping function include trifluoroacetic anhydride, formicacetic anhydride, or other reactive acylating agents. Finally, the slides are washed again with DMF, methylene chloride, and ethanol. [0550]
  • 6. Synthesis of Eight Trimers of C and T [0551]
  • FIG. 2 illustrates a possible synthesis of the eight trimers of the two-monomer set: cytosine and thymine (represented by C and T, respectively). A glass slide bearing silane groups terminating in 6-nitroveratryloxycarboxamide (NVoC-NH) residues is prepared as a substrate. Active esters (pentafluorophenyl, OBt, etc.) of cytosine and thymine protected at the 5′ hydroxyl group with NVOC are prepared as reagents. While not pertinent to this example, if side chain protecting groups are required for the monomer set, these must not be photoreactive at the wavelength of light used to protect the primary chain. [0552]
  • For a monomer set of size n, n×1 cycles are required to synthesize all possible sequences of length 1. A cycle consists of: [0553]
  • 1. Irradiation through an appropriate mask to expose the 5′-OH groups at the sites where the next residue is to be added, with appropriate washes to remove the by-products of the deprotection. [0554]
  • 2. Addition of a single activated and protected (with the same photochemically-removable group) monomer, which will react only at the sites addressed in step 1, with appropriate washes to remove the excess reagent from the surface. [0555]
  • The above cycle is repeated for each member of the monomer set until each location on the surface has been extended by one residue in one embodiment. In other embodiments, several residues are sequentially added at one location before moving on to the next location. Cycle times will generally be limited by the coupling reaction rate, now as short as about 10 min in automated oligonucleotide synthesizers. This step is optionally followed by addition of a protecting group to stabilize the array for later testing. For some types of polymers (e.g., peptides), a final deprotection of the entire surface (removal of photoprotective side chain groups) may be required. [0556]
  • More particularly, as shown in FIG. 2A, the [0557] glass 20 is provided with regions 22, 24, 26, 28, 30, 32, 34, and 36. Regions 30, 32, 34, and 36 are masked, indicated by the hatched regions, as shown in FIG. 2B and the glass is irradiated by the bright regions 22, 24, 26, and 28, and exposed to a reagent containing a photosensitive blocked C (e.g., cytosine derivative), with the resulting structure shown in FIG. 2C. The substrate is carefully washed and the reactants removed. Thereafter, regions 22, 24, 26, and 28 are masked, as indicated by the hatched region, the glass is irradiated (as shown in FIG. 2D), as indicated by the bright regions, at 30, 32, 34, and 36, and exposed to a photosensitive blocked reagent containing T (e.g., thymine derivative), with the resulting structure shown in FIG. 2E. The process proceeds, consecutively masking and exposing the sections as shown until the structure shown in FIG. 2M is obtained. The glass is irradiated and the terminal groups are, optionally, capped by acetylation. As shown, all possible trimers of cytosine/thymine are obtained.
  • In this example, no side chain protective group removal is necessary, as might be common in modified nucleotides. If it is desired, side chain deprotection may be accomplished by treatment with ethanedithiol and trifluoroacetic acid. [0558]
  • In general, the number of steps needed to obtain a particular polymer chain is defined by:[0559]
  • 1  (1)
  • where: [0560]
  • n=the number of monomers in the basis set of monomers, and [0561]
  • 1=the number of monomer units in a polymer chain. [0562]
  • Conversely, the synthesized number of sequences of length 1 will be:[0563]
  • n 1.  (2)
  • Of course, greater diversity is obtained by using masking strategies which will also include the synthesis of polymers having a length of less than 1. If, in the extreme case, all polymers having a length less than or equal to 1 are synthesized, the number of polymers synthesized will be:[0564]
  • n 1 +n 1−1 + . . . +n 1.  (3)
  • The maximum number of lithographic steps needed will generally be n for each “layer” of monomers, i.e., the total number of masks (and, therefore, the number of lithographic steps) needed will be n×1. The size of the transparent mask regions will vary in accordance with the area of the substrate available for synthesis and the number of sequences to be formed. In general, the size of the synthesis areas will be:[0565]
  • size of synthesis areas=(A)/(S)
  • where: [0566]
  • A is the total area available for synthesis; and [0567]
  • S is the number of sequences desired in the area. [0568]
  • It will be appreciated by those of skill in the art that the above method could readily be used to simultaneously produce thousands or millions of oligomers on a substrate using the photolithographic techniques disclosed herein. Consequently, the method results in the ability to practically test large numbers of, for example, di, tri, tetra, penta, hexa, hepta, octa, nona, deca, even dodecanucleotides, or larger polynucleotides (or correspondingly, polypeptides). [0569]
  • The above example has illustrated the method by way of a manual example. It will of course be appreciated that automated or semi-automated methods could be used. The substrate would be mounted in a flow cell for automated addition and removal of reagents, to minimize the volume of reagents needed, and to more carefully control reaction conditions. Successive masks will be applicable manually or automatically. See, e.g., Pirrung et al. (1992) U.S. Pat. No. 5,143,854 and Ser. No. 07/624,120, now abandoned. [0570]
  • 7. Labeling of Target [0571]
  • The target oligonucleotide can be labeled using standard procedures referred to above. As discussed, for certain situations, a reagent which recognizes interaction, e.g., ethidium bromide, may be provided in the detection step. Alternatively, fluorescence labeling techniques may be applied, see, e.g., Smith, et al. (1986) [0572] Nature, 321: 674-679; and Prober, et al. (1987) Science, 238:336-341. The techniques described therein will be followed with minimal modifications as appropriate for the label selected.
  • 8. Dimers of A, C, G, and T [0573]
  • The described technique may be applied, with photosensitive blocked nucleotides corresponding to adenine, cytosine, guanine, and thymine, to make combinations of polynucleotides consisting of each of the four different nucleotides. All 16 possible dimers would be made using a minor modification of the described method. [0574]
  • 9. 10-mers of A, C, G, and T [0575]
  • The described technique for making dimers of A, C, G, and T may be further extended to make longer oligonucleotides. The automated system described, e.g., in Pirrung et al. (1992) U.S. Pat. No. 5,143,854, and Ser. No. 07/624,120, now abandoned, can be adapted to make all possible 10-mers composed of the 4 nucleotides A, C, G, and T. The photosensitive, blocked nucleotide analogues have been described above, and would be readily adaptable to longer oligonucleotides. [0576]
  • 10. Specific Recognition Hybridization to 10-mers [0577]
  • The described hybridization conditions are directly applicable to the sequence specific recognition reagents attached to the substrate, produced as described immediately above. The 10-mers have an inherent property of hybridizing to a complementary sequence. For optimum discrimination between full matching and some mismatch, the conditions of hybridization should be carefully selected, as described above. Careful control of the conditions, and titration of parameters should be performed to determine the optimum collective conditions. [0578]
  • 11. Hybridization [0579]
  • Hybridization conditions are described in detail, e.g., in Hames and Higgins (1985) [0580] Nucleic Acid Hybridisation: A Practical Approach; and the considerations for selecting particular conditions are described, e.g., in Wetmur and Davidson, (1988) J. Mol. Biol. 31:349-370, and Wood et al. (1985) Proc. Natl. Acad. Sci. USA 82:1585-1588. As described above, conditions are desired which can distinguish matching along the entire length of the probe from where there is one or more mismatched bases. The length of incubation and conditions will be similar, in many respects, to the hybridization conditions used in Southern blot transfers. Typically, the GC bias may be minimized by the introduction of appropriate concentrations of the alkylammonium buffers, as described above.
  • Titration of the temperature and other parameters is desired to determine the optimum conditions for specificity and distinguishability of absolutely matched hybridization from mismatched hybridization. [0581]
  • A fluorescently labeled target or set of targets are generated, as described in Prober, et al. (1987) [0582] Science 238:336-341, or Smith, et al. (1986) Nature 321:674-679. Preferably, the target or targets are of the same length as, or slightly longer, than the oligonucleotide probes attached to the substrate and they will have known sequences. Thus, only a few of the probes hybridize perfectly with the target, and which particular ones did would be known.
  • The substrate and probes are incubated under appropriate conditions for a sufficient period of time to allow hybridization to completion. The time is measured to determine when the probe-target hybridizations have reached completion. A salt buffer which minimizes GC bias is preferred, incorporating, e.g., buffer, such as tetramethyl ammonium or tetraethyl ammonium ion at between about 2.4 and 3.0 M. See Wood, et al. (1985) [0583] Proc. Nat'l Acad. Sci. USA 82:1585-1588. This time is typically at least about 30 min, and may be as long as about 1-5 days. Typically very long matches will hybridize more quickly, very short matches will hybridize less quickly, depending upon relative target and probe concentrations. The hybridization will be performed under conditions where the reagents are stable for that time duration.
  • Upon maximal hybridization, the conditions for washing are titrated. Three parameters initially titrated are time, temperature, and cation concentration of the wash step. The matrix is scanned at various times to determine the conditions at which the distinguishability between true perfect hybrid and mismatched hybrid is optimized. These conditions will be preferred in the sequencing embodiments. [0584]
  • 12. Positional Detection of Specific Interaction [0585]
  • As indicated above, the detection of specific interactions may be performed by detecting the positions where the labeled target sequences are attached. Where the label is a fluorescent label, the apparatus described, e.g., in Pirrung et al. (1992) U.S. Pat. No. 5,143,854; and Ser. No. 07/624,120, now abandoned, may be advantageously applied. In particular, the synthetic processes described above will result in a matrix pattern of specific sequences attached to the substrate, and a known pattern of interactions can be converted to corresponding sequences. [0586]
  • In an alternative embodiment, a separate reagent which differentially interacts with the probe and interacted probe/targets can indicate where interaction occurs or does not occur. A single-strand specific reagent will indicate where no interaction has taken place, while a double-strand specific reagent will indicate where interaction has taken place. An intercalating dye, e.g., ethidium bromide, may be used to indicate the positions of specific interaction. [0587]
  • 13. Analysis [0588]
  • Conversion of the positional data into sequence specificity will provide the set of subsequences whose analysis by overlap segments, may be performed, as described above. Analysis is provided by the methodology described above, or using, e.g., software available from the Genetic Engineering Center, P.O. Box 794, 11000 Belgrade, Yugoslavia (Yugoslav group). See, also, Macevicz, PCT publication no. WO 90/04652, which is hereby incorporated herein by reference. [0589]
  • B. Polypeptide [0590]
  • The description of the preparation of short peptides on a substrate incorporates by reference sections in Pirrung et al. (1992) U.S. Pat. No. 5,143,854, and described below. [0591]
  • 1. Slide Preparation [0592]
  • Preparation of the substrate follows that described above for nucleotides. [0593]
  • 2. Linker Attachment, Blocking of Free Sites [0594]
  • The aminated surface of the slide is exposed to about 500 μl of, e.g., a 30 millimolar (mM) solution of NVOC-GABA (gamma amino butyric acid) NES (N-hydroxysuccinimide) in DMF for attachment of a NVOC-GABA to each of the amino groups. The surface is washed with, for example, DMF, methylene chloride, and ethanol. See Ser. No. 07,624,120, now abandoned, for details on amino acid chemistry. [0595]
  • Any unreacted aminopropyl silane on the surface, i.e., those amino groups which have not had the NVOC-GABA attached, are now capped with acetyl groups (to prevent further reaction) by exposure to a 1:3 mixture of acetic anhydride in pyridine for 1 hour. Other materials which may perform this residual capping function include trifluoroacetic anhydride, formicacetic anhydride, or other reactive acylating agents. Finally, the slides are washed again with DMF, methylene chloride, and ethanol. [0596]
  • 3. Synthesis of 8 Trimers of “A” and “B”[0597]
  • See Pirrung et al. (1992) U.S. Pat. No. 5,143,854 which describes the preparation of glycine and phenylalanine trimers. The technique is similar to the method described above for making triners of C and T, but substituting photosensitive blocked glycine for the C derivative and photosensitive blocked phenylalamine for the T derivative. [0598]
  • 4. Synthesis of a Dimer of an Aminopropyl Group and a Fluorescent Group [0599]
  • In synthesizing the dimer of an aminopropyl group and a fluorescent group, a functionalized Durapore™ membrane was used as a substrate. The Durapore™ membrane was a polyvinylidine difluoride with aminopropyl groups. The aminopropyl groups were protected with the DDZ group by reaction of the carbonyl chloride with the amino groups, a reaction readily known to those of skill in the art. The surface bearing these groups was placed in a solution of THF and contacted with a mask bearing a checkerboard pattern of 1 mm opaque and transparent regions. The mask was exposed to ultraviolet light having a wavelength down to at least about 280 nm for about 5 minutes at ambient temperature, although a wide range of exposure times and temperatures may be appropriate in various embodiments of the invention. For example, in one embodiment, an exposure time of between about 1 and 5000 seconds may be used at process temperatures of between −70 and +50° C. [0600]
  • In one preferred embodiment, exposure times of between about 1 and 500 seconds at about ambient pressure are used. In some preferred embodiments, pressure above ambient is used to prevent evaporation. [0601]
  • The surface of the membrane was then washed for about 1 hour with a fluorescent label which included an active ester bound to a chelate of a lanthanide. Wash times will vary over a wide range of values from about a few minutes to a few hours. These materials fluoresce in the red and the green visible region. After the reaction with the active ester in the fluorophore was complete, the locations in which the fluorophore was bound could be visualized by exposing them to ultraviolet light and observing the red and the green fluorescence. It was observed that the derivatized regions of the substrate closely corresponded to the original pattern of the mask. [0602]
  • 5. Demonstration of Signal Capability [0603]
  • Signal detection capability was demonstrated using a low-level standard fluorescent bead kit manufactured by Flow Cytometry Standards and having model no. 824. This kit includes 5.8 μm diameter beads, each impregnated with a known number of fluorescein molecules. [0604]
  • One of the beads was placed in the illumination field on the scan stage in a field of a laser spot which was initially shuttered. After being positioned in the illumination field, the photon detection equipment was turned on. The laser beam was unblocked and it interacted with the particle bead, which then fluoresced. Fluorescence curves of beads impregnated with 7,000 and 29,000 fluorescein molecules, are shown in FIGS. 11A and 11B, respectively of Pirrung et al. (1992) U.S. Pat. No. 5,143,854. On each curve, traces for beads without fluorescein molecules are also shown. These experiments were performed with 488 nm excitation, with 100 μW of laser power. The light was focused through a 40 power 0.75 NA objective. [0605]
  • The fluorescence intensity in all cases started off at a high value and then decreased exponentially. The fall-off in intensity is due to photobleaching of the fluorescein molecules. The traces of beads without fluorescein molecules are used for background subtraction. The difference in the initial exponential decay between labeled and nonlabeled beads is integrated to give the total number of photon counts, and this number is related to the number of molecules per bead. Therefore, it is possible to deduce the number of photons per fluorescein molecule that can be detected. This calculation indicates the radiation of about 40 to 50 photons per fluorescein molecule are detected. [0606]
  • 6. Determination of the Number of Molecules Per Unit Area [0607]
  • Aminopropylated glass microscope slides prepared according to the methods discussed above were utilized in order to establish the density of labeling of the slides. The free amino termini of the slides were reacted with FITC (fluorescein isothiocyanate) which forms a covalent linkage with the amino group. The slide is then scanned to count the number of fluorescent photons generated in a region which, using the estimated 40-50 photons per fluorescent molecule, enables the calculation of the number of molecules which are on the surface per unit area. [0608]
  • A slide with aminopropyl silane on its surface was immersed in a 1 mM solution of FITC in DMF for 1 hour at about ambient temperature. After reaction, the slide was washed twice with DMF and then washed with ethanol, water, and then ethanol again. It was then dried and stored in the dark until it was ready to be examined. [0609]
  • Through the use of curves similar to those shown in FIG. 11 of Pirrung et al. (1992) U.S. Pat. No. 5,143,854, and by integrating the fluorescent counts under the exponentially decaying signal, the number of free amino groups on the surface after derivitization was determined. It was determined that slides with labeling densities of 1 fluorescein per 10[0610] 3×103 to ˜2×2 nm could be reproducibly made as the concentration of aminopropyltriethoxysilane varied from 10−5% to 10−1%.
  • 7. Removal of NVOC and Attachment of a Fluorescent Marker [0611]
  • NVOC-GABA groups were attached as described above. The entire surface of one slide was exposed to light so as to expose a free amino group at the end of the gamma amino butyric acid. This slide, and a duplicate which was not exposed, were then exposed to fluorescein isothiocyanate (FITC). [0612]
  • FIG. 12A of Pirrung et al. (1992) U.S. Pat. No. 5,143,854 illustrates the slide which was not exposed to light, but which was exposed to FITC. The units of the x axis are time and the units of the y axis are counts. The trace contains a certain amount of background fluorescence. The duplicate slide was exposed to 350 nm broadband illumination for about 1 minute (12 mW/cm[0613] 2, ˜350 nm illumination) , washed and reacted with FITC. A large increase in the level of fluorescence is observed, which indicates photolysis has exposed a number of amino groups on the surface of the slides for attachment of a fluorescent marker.
  • 8. Use of a Mask in Removal of NVOC [0614]
  • The next experiment was performed with a 0.1% aminopropylated slide. Light from a Hg-Xe arc lamp was imaged onto the substrate through a laser-ablated chrome-on-glass mask in direct contact with the substrate. [0615]
  • This slide was illuminated for approximately 5 minutes, with 12 mW of 350 nm broadband light and then reacted with the 1 mM FITC solution. It was put on the laser detection scanning stage and a graph was plotted as a two-dimensional representation of position color-coded for fluorescence intensity. The experiment was repeated a number of times through various masks. The fluorescence patterns for a 100×100 μm mask, a 50 μm mask, a 20 μm mask, and a 10 μm mask indicate that the mask pattern is distinct down to at least about 10 μm squares using this lithographic technique. [0616]
  • 9. Attachment of YGGFL and Subsequent Exposure to Herz Antibody and Goat Anti-mouse Antibody [0617]
  • In order to establish that receptors to a particular polypeptide sequence would bind to a surface-bound peptide and be detected, Leu enkephalin was coupled to the surface and recognized by an antibody. A slide was derivatized with 0.1% amino propyl-triethoxysilane and protected with NVOC. A 500 μm checkerboard mask was used to expose the slide in a flow cell using backside contact printing. The Leu enkephalin sequence (H[0618] 2N-tyrosine,glycine,glycine,phenylalanine,leucine-COOH, otherwise referred to herein as YGGFL) was attached via its carboxy end to the exposed amino groups on the surface of the slide. The peptide was added in DMF solution with the BOP/HOBT/DIEA coupling reagents and recirculated through the flow cell for 2 hours at room temperature.
  • A first antibody, known as the Herz antibody, was applied to the surface of the slide for 45 minutes at 2 μg/ml in a supercocktail (containing 1% BSA and 1% ovalbumin also in this case) . A second antibody, goat anti-mouse fluorescein conjugate, was then added at 2 μg/ml in the supercocktail buffer, and allowed to incubate for 2 hours. [0619]
  • The results of this experiment were plotted as fluorescence intensity as a function of position. This image was taken at 10 μm steps and showed that not only can deprotection be carried out in a well defined pattern, but also that (1) the method provided for successful coupling of peptides to the surface of the substrate, (2) the surface of a bound peptide was available for binding with an antibody, and (3) the detection apparatus capabilities were sufficient to detect binding of a receptor. Moreover, the Herz antibody is a sequence specific reagent which may be used advantageously as a sequence specific recognition reagent. It may be used, if specificity is high, for sequencing purposes, and, at least, for fingerprinting and mapping uses. [0620]
  • 10. Monomer-by-monomer Formation of YGGFL and Subsequent Exposure to Labeled Antibody [0621]
  • Monomer-by-monomer synthesis of YGGFL and GGFL in alternate squares was performed on a slide in a checkerboard pattern and the resulting slide was exposed to the Herz antibody. [0622]
  • A slide is derivatized with the aminopropyl group, protected in this case with t-BOC (t-butoxycarbonyl). The slide was treated with TFA to remove the t-BOC protecting group. E-aminocaproic acid, which was t-BOC protected at its amino group, was then coupled onto the aminopropyl groups. The aminocaproic acid serves as a spacer between the aminopropyl group and the peptide to be synthesized. The amino end of the spacer was deprotected and coupled to NVOC-leucine. The entire slide was then illuminated with 12 mW of 325 nm broadband illumination. The slide was then coupled with NVOC-phenylalanine and washed. The entire slide was again illuminated, then coupled to NVOC-glycine and washed. The slide was again illuminated and coupled to NVOC-glycine to form the sequence shown in the last portion of FIG. 13A of Pirrung et al. (1992) U.S. Pat. No. 5,143,854. [0623]
  • Alternating regions of the slide were then illuminated using a projection print using a 500×500 μm checkerboard mask; thus, the amino group of glycine was exposed only in the lighted areas. When the next coupling chemistry step was carried out, NVOC-tyrosine was added, and it coupled only at those spots which had received illumination. The entire slide was then illuminated to remove all the NVOC groups, leaving a checkerboard of YGGFL in the lighted areas and in the other areas, GGFL. The Herz antibody (which recognizes the YGGFL, but not GGFL) was then added, followed by goat anti-mouse fluorescein conjugate. [0624]
  • The resulting fluorescence scan showed dark areas containing the tetrapeptide GGFL, which is not recognized by the Herz antibody (and thus there is no binding of the goat anti-mouse antibody with fluorescein conjugate), and red areas in which YGGFL was present. The YGGFL pentapeptide is recognized by the Herz antibody and, therefore, there is antibody in the lighted regions for the fluorescein-conjugated goat anti-mouse to recognize. [0625]
  • Similar patterns for a 50 μm mask used in direct contact (“proximity print”) with the substrate provided a pattern which was more distinct and the corners of the checkerboard pattern were touching as a result of the mask being placed in direct contact with the substrate (which reflects the increase in resolution using this technique). [0626]
  • 11. Monomer-by-monomer Synthesis of YGGFL and PGGFL [0627]
  • A synthesis using a 50 μm checkerboard mask was conducted. However, P was added to the GGFL sites on the substrate through an additional coupling step. P was added by exposing protected GGFL to light through a mask, and subsequence exposure to P in the manner set forth above. Therefore, half of the regions on the substrate contained YGGFL and the remaining half contained PGGFL. [0628]
  • The fluorescence plot for this experiment showed the regions are again readily discernable between those in which binding did and did not occur. This experiment demonstrated that antibodies are able to recognize a specific sequence and that the recognition is not length-dependent. [0629]
  • 12. Monomer-by-monomer Synthesis of YGGFL and YPGGFL [0630]
  • In order to further demonstrate the operability of the invention, a 50 μm checkerboard pattern of alternating YGGFL and YPGGFL was synthesized on a substrate using techniques like those set forth above. The resulting fluorescence plot showed that the antibody was clearly able to recognize the YGGFL sequence and did not bind significantly at the YPGGFL regions. [0631]
  • 13. Synthesis of an Array of Sixteen Different Amino Acid Sequences and Estimation of Relative Binding Affinity to Herz Antibody [0632]
  • Using techniques similar to those set forth above, an array of 16 different amino acid sequences (replicated four times) was synthesized on each of two glass substrates. The sequences were synthesized by attaching the sequence NVOC-GFL across the entire surface of the slides. Using a series of masks, two layers of amino acids were then selectively applied to the substrate. Each region had dimensions of 0.25 cm×0.0625 cm. The first slide contained amino acid sequences containing only L- amino acids while the second slide contained selected D- amino acids. Various regions on the first and second slides, were duplicated four times on each slide. The slides were then exposed to the Herz antibody and fluorescein-labeled goat anti-mouse antibodies. [0633]
  • A fluorescence plot of the first slide, which contained only L- amino acids showed red areas (indicating strong binding, i.e., 149,000 counts or more) and black areas (indicating little or no binding of the Herz antibody, i.e., 20,000 counts or less). The sequence YGGFL was clearly most strongly recognized. The sequences YAGFL and YSGFL also exhibited strong recognition of the antibody. By contrast, most of the remaining sequences showed little or no binding. The four duplicate portions of the slide were extremely consistent in the amount of binding shown therein. [0634]
  • A fluorescence plot of the D- amino acid slide indicated that strongest binding was exhibited by the YGGFL sequence. significant binding was also detected to YaGFL, YsGFL, and YpGFL. The remaining sequences showed less binding with the antibody. Low binding efficiency of the sequence yGGFL was observed. [0635]
  • Table 6 lists the various sequences tested in order of relative fluorescence, which provides information regarding relative binding affinity. [0636]
    TABLE 6
    Apparent Binding to Herz Ab
    L- a.a. Set D- a.a. Set
    YGGFL YGGFL
    YAGFL YaGFL
    YSGFL YsGFL
    LGGFL YpGFL
    FGGFL fGGFL
    YPGFL yGGFL
    LAGFL faGFL
    FAGFL wGGFL
    WGGFL yaGFL
    fpGFL
    waGFL
  • 14. Illustrative Alternative Embodiment [0637]
  • According to an alternative embodiment of the invention, the methods provide for attaching to the surface a caged binding member which, in its caged form, has a relatively low affinity for other potentially binding species, such as receptors and specific binding substances. Such techniques are more fully described in copending application Ser. No. 404,920, filed Sep. 8, 1989, and incorporated herein by reference for all purposes. See also Ser. No. 07/435,316, now abandoned, and Barrett et al. (1993) U.S. Pat. No. 5,252,743, each of which is hereby incorporated herein by reference. [0638]
  • According to this alternative embodiment, the invention provides methods for forming predefined regions on a surface of a solid support, wherein the predefined regions are capable of immobilizing receptors. The methods make use of caged binding members attached to the surface to enable selective activation of the predefined regions. The caged binding members are liberated to act as binding members ultimately capable of binding receptors upon selective activation of the predefined regions. The activated binding members are then used to immobilize specific molecules such as receptors on the predefined region of the surface. The above procedure is repeated at the same or different sites on the surface so as to provide a surface prepared with a plurality of regions on the surface containing, for example, the same or different receptors. When receptors immobilized in this way have a differential affinity for one or more ligands, screenings and assays for the ligands can be conducted in the regions of the surface containing the receptors. [0639]
  • The alternative embodiment may make use of novel caged binding members attached to the substrate. Caged (unactivated) members have a relatively low affinity for receptors of substances that specifically bind to uncaged binding members when compared with the corresponding affinities of activated binding members. Thus, the binding members are protected from reaction until a suitable source of energy is applied to the regions of the surface desired to be activated. Upon application of a suitable energy source, the caging groups labilize, thereby presenting the activated binding member. A typical energy source will be light. [0640]
  • Once the binding members on the surface are activated they may be attached to a receptor. The receptor chosen may be a monoclonal antibody, a nucleic acid sequence, a drug receptor, etc. The receptor will usually, though not always, be prepared so as to permit attaching it, directly or indirectly, to a binding member. For example, a specific binding substance having a strong binding affinity for the binding member and a strong affinity for the receptor or a conjugate of the receptor may be used to act as a bridge between binding members and receptors if desired. The method uses a receptor prepared such that the receptor retains its activity toward a particular ligand. [0641]
  • Preferably, the caged binding member attached to the solid substrate will be a photoactivatable biotin complex, i.e., a biotin molecule that has been chemically modified with photoactivatable protecting groups so that it has a significantly reduced binding affinity for avidin or avidin analogs than does natural biotin. In a preferred embodiment, the protecting groups localized in a predefined region of the surface will be removed upon application of a suitable source of radiation to give binding members, that is biotin or a functionally analogous compound having substantially the same binding affinity for avidin or avidin analogs as does biotin. [0642]
  • In another preferred embodiment, avidin or an avidin analog is incubated with activated binding members on the surface until the avidin binds strongly to the binding members The avidin so immobilized on predefined regions of the surface can then be incubated with a desired receptor or conjugate of a desired receptor. The receptor will preferably be biotinylated, e.g., a biotinylated antibody, when avidin is immobilized on the predefined regions of the surface. Alternatively, a preferred embodiment will present an avidin/biotinylated receptor complex, which has been previously prepared, to activated binding members on the surface. [0643]
  • II. Fingerprinting [0644]
  • The above section on generation of reagents for sequencing provides specific reagents useful for fingerprinting applications. Fingerprinting embodiments may be applied towards polynucleotide fingerprinting, polypeptide fingerprinting, cell and tissue classification, cell and tissue temporal development stage classification, diagnostic tests, forensic uses for individual identification, classification of organisms, and genetic screening of individuals. Mapping applications are also described below. [0645]
  • A. Polynucleotide Fingerprint [0646]
  • Polynucleotide fingerprinting may use reagents similar to those described above for probing a sequence for the presence of specific subsequences found therein. Typically, the subsequences used for fingerprinting will be longer than the sequences used in oligonucleotide sequencing. In particular, specific long segments may be used to determine the similarity of different samples of nucleic acids. They may also be used to fingerprint whether specific combinations of information are provided therein. Particular probe sequences are selected and attached in a positional manner to a substrate. The means for attachment may be either using a caged biotin method described, e.g., in Barrett et al. (1993) U.S. Pat. No. 5,242,743, or by another method using targeting molecules. For example, a short polypeptide of specific sequence may be attached to an oligonucleotide and targeted to specific positions on a substrate having antibodies attached thereto, the antibodies exhibiting specificity for binding to those short peptide sequences. In another embodiment, an unnatural nucleotide or similar complementary binding molecule may be attached to the fingerprinting probe and the probe thereby directed towards complementary sequences on a VLSIPS substrate. Typically, unnatural nucleotides would be preferred, e.g., unnatural optical isomers, which would not interfere with natural nucleotide interactions. [0647]
  • Having produced a substrate with particular fingerprint probes attached thereto at positionally defined regions, the substrate may be used in a manner quite similar to the sequencing embodiment to provide information as to whether the fingerprint probes are detecting the corresponding sequence in a target sequence. This will often provide information similar to a Southern blot hybridization. [0648]
  • B. Polypeptide Fingerprint [0649]
  • A polypeptide fingerprint may be performed using antibodies which recognize specific antigens on the polypeptide. For example, monoclonal antibodies which recognize specific sequences or antigens on a polypeptide may be used to determine whether those epitopes are found on a particular protein. For example, particular patterns of epitopes would be found on various types of proteins. This will lead to the discovery that specific epitopes, or antigenic determinants, which are characteristic of, e.g., beta sheet segments, will be identified as will particular different types of domains in various protein types. Thus, a screening method may be devised which can classify polypeptides, either native or denatured, into various new classes defined by the epitopes existing thereon. [0650]
  • In addition, once the substrate is generated in the manners described above, a target peptide is exposed to the substrate. The target may be either native or denatured, though the conditions used to denature the polypeptide may interfere with the specific interaction between the polypeptide and the recognition reagent. This method is not dependent on the fact that the polypeptide is a single chain, thus protein complexes may also be fingerprinted using this methodology. Structures such as multi-subunit proteins, associations of proteins, ribosomes, nucleosomes, and other small cellular structures may also be fingerprinted and classified according to the presence of specific recognizable features thereon. [0651]
  • Peptide fingerprinting may be useful, for example, in correlating with particular physiological conditions or developmental stages of a cell or organism. Thus, a biological sample may be fingerprinted to determine the presence in that sample of a plurality of different polypeptides which are each individually fingerprinted. In an alternative embodiment, a polypeptide itself is not fingerprinted but a biological sample is fingerprinted searching for specific epitopes, e.g., polypeptide, carbohydrate, nucleic acid, or any of a number of other specific recognizable structural features. [0652]
  • The conditions for the interactions using antibodies is described, e.g., in Harlow and Lane (1988) [0653] Antibodies: A Laboratory Manual, Cold Spring Harbor Press, New York. The conditions should be titrated for temperature, buffer composition, time, and other important parameters in an antibody interaction.
  • C. Cell Classification Scheme [0654]
  • The present invention can be used for cell classification using fingerprinting type technology as described above in the polypeptide fingerprint. Classes of cells are typically defined by the presence of common functions which are usually reflected by structural features. Thus, a plant cell is classified differently from an animal cell by a number of structural features. Given an unknown cell, the present invention provides improved means for distinguishing the different cell types. Once a cell classification scheme is developed and the structural features which define it are identified using the present invention, homogeneous cell population expressing these features may be separated from others. Standard cell sorters may be coupled with recognition reagents and labels which can distinguish various cell types. [0655]
  • a. T-Cell Classes [0656]
  • T-cell classes are defined on the basis of expression of particular antigens characteristic of each class. For example, mouse T-cell differentiation markers include the LY antigens. With the plurality of different antigens which may be tested using antibody or other recognition reagents, new populations and classes of cells may be defined. For example, different neural cell types may be defined on the basis of cell surface antigens. Different tissue types will be defined on the basis of tissue specific antigens. Developmental cell classes will be similarly defined. All of these screenings can make use of the VLSIPS substrates with specific recognition molecules attached thereto. The substrates are exposed to the cell types directly, assaying for attachment of cells to specific regions, or are exposed to products of a population of cells, e.g., a supernatant, or a cell lysate. [0657]
  • Once a cell classification scheme has been correlated with specific structural markers therein, reagents which recognize those features may be developed and used in a fluorescence activated cell sorter as described, e.g., in Dangl, J. and Herzenberg (1982) [0658] J. Immunological Methods 52: 1-14; and Becton Dickinson, Fluorescence Activated Cell Sorters Division, San Jose, Calif. This will provide a homogeneous population of cells whose function has been defined by structure.
  • b. B-Cell Classes [0659]
  • The present cell classification scheme may also be used to determine specific B-cell classes. For example, B-cells specific for producing IgM, IgG, IgD, IgE, and IgA may be defined by the internal expression of specific mRNA sequences encoding each type of immunoglobulin. The classification scheme may depend on either extracellularly expressed markers which are correlated as being diagnostic of specific stages in development, or intracellular mRNA sequences which indicate particular functions. [0660]
  • D. Temporal Development Scheme [0661]
  • 1. Developmental Antigens [0662]
  • The present fingerprinting invention also allows cell classification by expression of developmental antigens. For example, a lymphocyte stem cell expresses a particular combination of antigens. As the lymphocyte develops through a program developmental scheme, at various stages it expresses particular antigens which are diagnostic of particular stages in development. Again, the fingerprinting methodology allows for the definition of specific structural features which are diagnostic of developmental or functional features which will allow classification of cells into temporal developmental classes. Cells, products of those cells, or lysates of those cells will be assayed to determine the developmental stage of the source cells. In this manner, once a developmental stage is defined, specific synchronized populations of cells will be selected out of another population. These synchronized populations may be very important in determining the biological mechanisms of development. [0663]
  • 2. Developmental mRNA Expression [0664]
  • Besides expressed antigens, the present invention also allows for fingerprinting of the mRNA population of a cell. In this fashion, the mRNA population, which should be a good determinant of developmental stage, will be correlated with other structural features of the cell. In this manner, cells at specific developmental stages will be characterized by the intracellular environment, as well as the extracellular environment. The present invention also allows the combination of definitions based, in part, upon antigens and, in part, upon mRNA expression. [0665]
  • In one embodiment, the two may be combined in a single incubation step. A particular incubation condition may be found which is compatible with both hybridization recognition and non-hybridization recognition molecules. Thus, e.g., an incubation condition may be selected which allows both specificity of antibody binding and specificity of nucleic acid hybridization. This allows simultaneous performance of both types of interactions on a single matrix. Again, where developmental mRNA patterns are correlated with structural features, or with probes which are able to hybridize to intracellular MRNA populations, a cell sorter may be used to sort specifically those cells having desired mRNA population patterns. [0666]
  • E. Diagnostic Tests [0667]
  • The present invention also provides the ability to perform diagnostic tests. Diagnostic tests typically are based upon a fingerprint type assay, which tests for the presence of specific diagnostic structural features. Thus, the present invention provides means for viral strain identification, bacterial strain identification, and other diagnostic tests using positionally defined specific reagents. The present invention also allows for determining a spectrum of allergies, diagnosing a biological sample for any or all of the above, and testing for many other conditions. [0668]
  • 1. Viral Identification [0669]
  • The present invention provides reagents and methodology for identifying viral strains. The specific reagents may be either antibodies or recognition proteins which bind to specific viral epitopes preferably surface exposed, but may make use of internal epitopes, e.g., in a denatured viral sample. In an alternative embodiment, the viral genome may be probed for specific sequences which are characteristic of particular viral strains. As above, a combination of the two may be performed simultaneously in a single interaction step, or in separate tests, e.g., for both genetic characteristics and epitope characteristics. [0670]
  • 2. Bacterial Identification [0671]
  • Similar techniques will be applicable to identifying a bacterial source. This may be useful in diagnosing bacterial infections, or in classifying sources of particular bacterial species. For example, the bacterial assay may be useful in determining the natural range of survivability of particular strains of bacteria across regions of the country or in different ecological niches. [0672]
  • 3. Other Microbiological Identifications [0673]
  • The present invention provides means for diagnosis of other microbiological and other species, e.g., protozoal species and parasitic species in a biological sample, but also provides the means for assaying a combination of different infections. For example, a biological specimen may be assayed for the presence of any or all of these microbiological species. In human diagnostic uses, typical samples will be blood, sputum, stool, urine, or other samples. [0674]
  • 4. Allergy Tests [0675]
  • An immobilized set of antigens may be attached to a solid substrate and, instead of the standard skin reaction tests, a blood sample may be assayed on such a substrate to determine the presence of antibodies, e.g., IgE or other type antibodies, which may be diagnostic of an allergic or immunological susceptibility. A standard radioallergosorbent test (RAST) may be used to check a much larger population of antigens. [0676]
  • In addition, an allergy like test may be used to diagnose the immunological history of a particular individual. For example, by testing the circulating antibodies in a blood sample, which reflects the immunological history and memory of an individual, it may be determined what infections may not have been historically presented to the immune system. In this manner, it may be possible to specifically supplement an immune system for a short period of time with IgG fractions made up of specific types of gamma globulins. Thus, hepatitis gamma globulin injections may be better designed for a particular environment to which a person is expected to be exposed. This also provides the ability to identify genetically equivalent individuals who have immunologically different experiences. Thus, a blood sample from an individual who has a particular combination of circulating antibodies will likely be different from the combination of circulating antibodies found in a genetically similar or identical individual. This could allow for the distinction between clones of particular animals, e.g., mice, rats, or other animals. [0677]
  • F. Individual Identification [0678]
  • The present invention provides the ability to fingerprint and identify a genetic individual. This individual may be a bacterial or lower microorganism, as described above in diagnostic tests, or of a plant or animal. An individual may be identified genetically or immunologically, as described. [0679]
  • 1. Genetic [0680]
  • Genetic fingerprinting has been utilized in comparing different related species in Southern hybridization blots. Genetic fingerprinting has also been used in forensic studies, see, e.g., Morris et al. (1989) [0681] J. Forensic Science 34: 1311-1317, and references cited therein. As described above, an individual may be identified genetically by a sufficiently large number of probes. The likelihood that another individual would have an identical pattern over a sufficiently large number of probes may be statistically negligible. However, it is often quite important that a large number of probes be used where the statistical probability of matching is desired to be particularly low. In fact, the probes will optimally be selected for having high heterogeneity among the population. In addition, the fingerprint method may make use of the pattern of homologies indicated by a series of more and more stringent washes. Then, each position has both a sequence specificity and a homology measurement, the combination of which greatly increases the number of dimensions and the statistical likelihood of a perfect pattern match with another genetic individual.
  • 2. Immunological [0682]
  • As indicated above in the diagnostic tests, it is possible to identify a particular immune system within a genetically homogeneous class of organisms by virtue of their immunological history. For example, a large colony of cloned mice may be distinguishable by virtue of each immunological history. For example, one mouse may have had an immunological response to exposure to antigen A to which her genetically identical sibling may have not been exposed. By virtue of this differential history, the first of the pair will likely have a high antibody titer against the antigen A whereas her genetically identical sibling will have not had a response to that antigen by virtue of never having been exposed to it. For this reason, immune systems may be identified by their immunological memories. Thus, immunological experience may also be a means for identifying a particular individual at a particular moment in her lifetime. [0683]
  • This same immunological screening may be used for other sorts of identifiable biological products. For example, an individual may be identified by her combination of expressed proteins. These proteins may reflect a physiological state of the individual, and would thus be useful in certain circumstances where diagnostic tests may be performed. For example, an individual may be identified, in part, by the presence of particular metabolic products. [0684]
  • In fact, a plant origin may be determined by virtue of having within its genome an unnatural sequence introduced to it by genetic breeders. Thus, a marker nucleic acid sequence may be introduced as a means to determine whether a genetic strain of a plant or animal originated from another particular source. [0685]
  • G. Genetic Screening [0686]
  • 1. Test Alleles with Markers [0687]
  • The present invention provides for the ability to screen for genetic variations of individuals. For example, a number of genetic diseases are linked with specific alleles. See, e.g., Scriber, C. et al. (eds.) (1989) [0688] The Metabolic Bases of Inherited Disease, McGraw-Hill, New York. In one embodiment, cystic fibrosis has been correlated with a specific gene, see, Gregory et al. (1990) Nature 347: 382-386. A number of alleles are correlated with specific genetic deficiencies. See, e.g., McKusick, V. (1990) Genetic Inheritance in Man: Catalogs of Autosomal Dominant, Autosomal Recessive, and X-linked Phenotypes, Johns Hopkins University Press, Baltimore; Ott, J. (1985) Analysis of Human Genetic Linkage, Johns Hopkins University Press, Baltimore; Track, R. et al. (1989) Banbury Report 32: DNA Technology and Forensic Science, Cold Spring Harbor Press, New York; each of which is hereby incorporated herein by reference.
  • 2. Amniocentesis [0689]
  • Typically, amniocentesis is used to determine whether chromosome translocations have occurred. The mapping procedure may provide the means for determining whether these translocations have occurred, and for detecting particular alleles of various markers. [0690]
  • III. Mapping [0691]
  • A. Positionally Located Clones [0692]
  • The present invention allows for the positional location of specific clones useful for mapping. For example, caged biotin may be used for specifically positioning a probe to a location on a matrix pattern. [0693]
  • In addition, the specific probes may be positionally directed to specific locations on a substrate by targeting. For example, polypeptide specific recognition reagents may be attached to oligonucleotide sequences which can be complementarily targeted to specific locations on a VLSIPS™ Technology substrate. Hybridization conditions, as applied for oligonucleotide probes, will be used to target the reagents to locations on a substrate having complementary oligonucleotides synthesized thereon. In another embodiment, oligonucleotide probes may be attached to specific polypeptide targeting reagents such as an antigen or antibody. These reagents can be directed towards a complementary antigen or antibody already attached to a VLSIPS substrate. [0694]
  • In another embodiment, an unnatural nucleotide which does not interfere with natural nucleotide complementary hybridization may be used to target oligonucleotides to particular positions on a substrate. Unnatural optical isomers of natural nucleotides should be ideal candidates. [0695]
  • In this way, short probes may be used to determine the mapping of long targets or long targets may be used to map the position of shorter probes. See, e.g., Craig et al. 1990 [0696] Nuc. Acids Res. 18: 2653-2660.
  • B. Positionally Defined Clones [0697]
  • Positionally defined clones may be transferred to a new substrate by either physical transfer or by synthetic means. Synthetic means may involve either a production of the probe on the substrate using the VLSIPS™ Technology synthetic methods, or may involve the attachment of a targeting sequence made by VLSIPS synthetic methods which will target that positionally defined clone to a position on a new substrate. Both methods will provide a substrate having a number of positionally defined probes useful in mapping. [0698]
  • IX. Conclusion [0699]
  • The present inventions provide greatly improved methods and apparatus for synthesis of polymers on substrates. It is to be understood that the above description is intended to be illustrative and not restrictive. Many embodiments will be apparent to those of skill in the art upon reviewing the above description. By way of example, the invention has been described primarily with reference to the use of photoremovable protective groups, but it will be readily recognized by those of skill in the art that sources of radiation other than light could also be used. For example, in some embodiments it may be desirable to use protective groups which are sensitive to electron beam irradiation, x-ray irradiation, in combination with electron beam lithograph, or x-ray lithography techniques. Alternatively, the group could be removed by exposure to an electric current. The scope of the invention should, therefore, be determined not with reference to the above description, but should instead be determined with reference to the appended claims, along with the full scope of equivalents to which such claims are entitled. [0700]
  • All publications and patent applications referred to herein are incorporated by reference to the same extent as if each individual publication or patent application was specifically and individually incorporated by reference. The present invention now being fully described, it will be apparent to one of ordinary skill in the art that many changes and modifications can be made thereto without departing from the spirit or scope of the appended claims. [0701]

Claims (25)

What is claimed is:
1. A composition comprising a plurality of positionally distinguishable sequence specific reagents attached to a solid substrate, which reagents are capable of specifically binding to a predetermined subunit sequence of a preselected multi-subunit length having at least three subunits, said reagents representing substantially all possible sequences of said preselected length.
2. A composition of claim 1, wherein said subunit sequence is a polynucleotide or a polypeptide.
3. A composition of claim 1, wherein said preselected multi-subunit length is five subunits and said subunit sequence is a polynucleotide sequence.
4. A composition of claim 1, wherein said specific reagent is an oligonucleotide of at least about five nucleotides.
5. A composition of claim 1, wherein said specific reagent is a monoclonal antibody.
6. A composition of claim 1, wherein said specific reagents are all attached to a single solid substrate.
7. A composition of claim 1, wherein said reagents comprise about 3000 different sequences.
8. A composition of claim 1, wherein said reagents represents at least about 25% of the possible subsequences of said preselected length.
9. A composition of claim 1, wherein said reagents are localized in regions of the substrate having a density of at least 25 regions per square centimeter.
10. A composition of claim 6, wherein said substrate has a surface area of less than about 4 square centimeters.
11. A method of analyzing a sequence of a polynucleotide or a polypeptide, said method comprising the step of:
a) exposing said polynucleotide or polypeptide to a composition of claim 1.
12. A method of identifying or comparing a target sequence with a reference, said method comprising the step of:
a) exposing said target sequence to a composition of claim 1;
b) determining the pattern of positions of said reagents which specifically interact with said target sequence; and
c) comparing said pattern with the pattern exhibited by said reference when exposed to said composition.
13. A method for sequencing a segment of a polynucleotide comprising the steps of:
a) combining:
i) a substrate comprising a plurality of chemically synthesized and positionally distinguishable oligonucleotides capable of recognizing defined oligonucleotide sequences; and
ii) a target polynucleotide; thereby forming high fidelity matched duplex structures of complementary subsequences of known sequence; and
b) determining which of said reagents have specifically interacted with subsequences in said target polynucleotide.
14. A method of claim 13, wherein said segment is substantially the entire length of said polynucleotide.
15. A method for sequencing a polymer, said method comprising the steps of:
a) preparing a plurality of reagents which each specifically bind to a subsequence of preselected length;
b) positionally attaching each of said reagents to one or more solid phase substrates, thereby producing substrates of positionally definable sequence specific probes;
c) combining said substrates with a target polymer whose sequence is to be determined; and
d) determining which of said reagents have specifically interacted with subsequences in said target polymer.
16. A method of claim 15, wherein said substrates are beads.
17. A method of claim 15, wherein said plurality of reagents comprise substantially all possible subsequences of said preselected length found in said target.
18. A method of claim 15, wherein said solid phase substrates are a single substrate having attached thereto reagents recognizing substantially all possible subsequences of preselected length found in said target.
19. A method of claim 15, further comprising the step of analyzing a plurality of said recognized subsequences to assemble a sequence of said target polymer.
20. A method of claim 16, wherein at least some of said plurality of substrates have one subsequence specific reagent attached thereto, and said substrates are coded to indicate the specificity of said reagent.
21. A method of using a fluorescent nucleotide to detect interactions with oligonucleotide probes of known sequence, said method comprising:
a) attaching said nucleotide to a target unknown polynucleotide sequence, and
b) exposing said target polynucleotide sequence to a collection of positionally defined oligonucleotide probes of known sequences to determine the sequences of said probes which interact with said target.
22. A method of claim 21, further comprising the step of:
a) collating said known sequences to determine the overlaps of said known sequences to determine the sequence of said target sequence.
23. A method of mapping a plurality of sequences relative to one another, said method comprising:
a) preparing a substrate having a plurality of positionally attached sequence specific probes;
b) exposing each of said sequences to said substrate, thereby determining the patterns of interaction between said sequence specific probes and said sequences; and
c) determining the relative locations of said sequence specific probe interactions on said sequences to determine the overlaps and order of said sequences.
24. A method of claim 23, wherein said sequence specific probes are oligonucleotides.
25. A method of claim 23, wherein said sequences are nucleic acid sequences.
US10/125,428 1990-12-06 2002-04-19 Arrays for detecting nucleic acids Abandoned US20020155491A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/125,428 US20020155491A1 (en) 1990-12-06 2002-04-19 Arrays for detecting nucleic acids

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US62411490A 1990-12-06 1990-12-06
US16890493A 1993-12-15 1993-12-15
US08/670,118 US5800992A (en) 1989-06-07 1996-06-25 Method of detecting nucleic acids
US09/056,927 US6197506B1 (en) 1989-06-07 1998-04-08 Method of detecting nucleic acids
US09/362,089 US6440667B1 (en) 1989-06-07 1999-07-28 Analysis of target molecules using an encoding system
US09/670,563 US6451536B1 (en) 1990-12-06 2000-09-27 Products for detecting nucleic acids
US09/768,626 US6576424B2 (en) 1989-06-07 2001-01-25 Arrays and methods for detecting nucleic acids
US10/125,428 US20020155491A1 (en) 1990-12-06 2002-04-19 Arrays for detecting nucleic acids

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/768,626 Continuation US6576424B2 (en) 1989-06-07 2001-01-25 Arrays and methods for detecting nucleic acids

Publications (1)

Publication Number Publication Date
US20020155491A1 true US20020155491A1 (en) 2002-10-24

Family

ID=24500711

Family Applications (15)

Application Number Title Priority Date Filing Date
US09/585,556 Expired - Fee Related US6544739B1 (en) 1990-12-06 2000-06-02 Method for marking samples
US09/670,563 Expired - Fee Related US6451536B1 (en) 1989-06-07 2000-09-27 Products for detecting nucleic acids
US09/768,626 Expired - Fee Related US6576424B2 (en) 1989-06-07 2001-01-25 Arrays and methods for detecting nucleic acids
US10/098,485 Abandoned US20030104411A1 (en) 1990-12-06 2002-03-18 Arrays for detecting nucleic acids
US10/098,484 Abandoned US20030119011A1 (en) 1989-06-07 2002-03-18 Arrays for detecting nucleic acids
US10/102,774 Abandoned US20050009014A9 (en) 1989-06-07 2002-03-22 Arrays for detecting nucleic acids
US10/102,915 Abandoned US20050170340A9 (en) 1989-06-07 2002-03-22 Arrays for detecting nucleic acids
US10/125,460 Abandoned US20020155492A1 (en) 1990-12-06 2002-04-19 Arrays for detecting nucleic acids
US10/125,530 Abandoned US20050053928A9 (en) 1990-03-07 2002-04-19 Arrays for detecting nucleic acids
US10/125,428 Abandoned US20020155491A1 (en) 1990-12-06 2002-04-19 Arrays for detecting nucleic acids
US10/694,541 Abandoned US20040067521A1 (en) 1990-12-06 2003-10-28 Arrays for detecting nucleic acids
US10/694,536 Abandoned US20040248147A1 (en) 1990-12-06 2003-10-28 Arrays for detecting nucleic acids
US10/992,772 Abandoned US20050164249A1 (en) 1990-12-06 2004-11-22 Arrays for detecting nucleic acids
US10/992,782 Abandoned US20050158743A1 (en) 1990-12-06 2004-11-22 Arrays for detecting nucleic acids
US10/993,432 Abandoned US20050112676A1 (en) 1990-12-06 2004-11-22 Arrays for detecting nucleic acids

Family Applications Before (9)

Application Number Title Priority Date Filing Date
US09/585,556 Expired - Fee Related US6544739B1 (en) 1990-12-06 2000-06-02 Method for marking samples
US09/670,563 Expired - Fee Related US6451536B1 (en) 1989-06-07 2000-09-27 Products for detecting nucleic acids
US09/768,626 Expired - Fee Related US6576424B2 (en) 1989-06-07 2001-01-25 Arrays and methods for detecting nucleic acids
US10/098,485 Abandoned US20030104411A1 (en) 1990-12-06 2002-03-18 Arrays for detecting nucleic acids
US10/098,484 Abandoned US20030119011A1 (en) 1989-06-07 2002-03-18 Arrays for detecting nucleic acids
US10/102,774 Abandoned US20050009014A9 (en) 1989-06-07 2002-03-22 Arrays for detecting nucleic acids
US10/102,915 Abandoned US20050170340A9 (en) 1989-06-07 2002-03-22 Arrays for detecting nucleic acids
US10/125,460 Abandoned US20020155492A1 (en) 1990-12-06 2002-04-19 Arrays for detecting nucleic acids
US10/125,530 Abandoned US20050053928A9 (en) 1990-03-07 2002-04-19 Arrays for detecting nucleic acids

Family Applications After (5)

Application Number Title Priority Date Filing Date
US10/694,541 Abandoned US20040067521A1 (en) 1990-12-06 2003-10-28 Arrays for detecting nucleic acids
US10/694,536 Abandoned US20040248147A1 (en) 1990-12-06 2003-10-28 Arrays for detecting nucleic acids
US10/992,772 Abandoned US20050164249A1 (en) 1990-12-06 2004-11-22 Arrays for detecting nucleic acids
US10/992,782 Abandoned US20050158743A1 (en) 1990-12-06 2004-11-22 Arrays for detecting nucleic acids
US10/993,432 Abandoned US20050112676A1 (en) 1990-12-06 2004-11-22 Arrays for detecting nucleic acids

Country Status (7)

Country Link
US (15) US6544739B1 (en)
EP (4) EP1231282A3 (en)
AU (1) AU1248292A (en)
DE (2) DE69132843T2 (en)
DK (2) DK0834576T3 (en)
HK (2) HK1010218A1 (en)
WO (1) WO1992010588A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020192684A1 (en) * 1989-06-07 2002-12-19 Affymetrix, Inc. Arrays for detecting nucleic acids
US20080286786A1 (en) * 2003-11-10 2008-11-20 Investigen, Inc. Methods of preparing nucleic acid for detection

Families Citing this family (414)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6270961B1 (en) * 1987-04-01 2001-08-07 Hyseq, Inc. Methods and apparatus for DNA sequencing and DNA identification
US5925525A (en) * 1989-06-07 1999-07-20 Affymetrix, Inc. Method of identifying nucleotide differences
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
US5871928A (en) * 1989-06-07 1999-02-16 Fodor; Stephen P. A. Methods for nucleic acid analysis
US5547839A (en) * 1989-06-07 1996-08-20 Affymax Technologies N.V. Sequencing of surface immobilized polymers utilizing microflourescence detection
US5800992A (en) * 1989-06-07 1998-09-01 Fodor; Stephen P.A. Method of detecting nucleic acids
US5846708A (en) * 1991-11-19 1998-12-08 Massachusetts Institiute Of Technology Optical and electrical methods and apparatus for molecule detection
EP1086742B2 (en) 1991-11-22 2007-03-14 Affymetrix, Inc. (a Delaware Corporation) Combinatorial strategies for polymer synthesis
US6943034B1 (en) 1991-11-22 2005-09-13 Affymetrix, Inc. Combinatorial strategies for polymer synthesis
US5565324A (en) * 1992-10-01 1996-10-15 The Trustees Of Columbia University In The City Of New York Complex combinatorial chemical libraries encoded with tags
US6503759B1 (en) 1992-10-01 2003-01-07 The Trustees Of Columbia University In The City Of New York Complex combinatorial chemical libraries encoded with tags
US5721099A (en) * 1992-10-01 1998-02-24 Trustees Of Columbia University In The City Of New York Complex combinatorial chemical libraries encoded with tags
US5795714A (en) * 1992-11-06 1998-08-18 Trustees Of Boston University Method for replicating an array of nucleic acid probes
EP0905258A3 (en) 1992-11-27 2001-01-31 Innogenetics N.V. Method for detecting nucleic acid sequences based on the use of solid phase immobilised nucleotide probes (line probe assay)
US7258977B1 (en) 1992-11-27 2007-08-21 Innogenetics N.V. Process for typing of HCV isolates
SE501439C2 (en) * 1993-06-22 1995-02-13 Pharmacia Lkb Biotech Method and apparatus for analyzing polynucleotide sequences
US7001722B1 (en) 1993-06-22 2006-02-21 Baylor College Of Medicine Parallel primer extension approach to nucleic acid sequence analysis
US6153379A (en) * 1993-06-22 2000-11-28 Baylor College Of Medicine Parallel primer extension approach to nucleic acid sequence analysis
WO1995011995A1 (en) * 1993-10-26 1995-05-04 Affymax Technologies N.V. Arrays of nucleic acid probes on biological chips
US6027880A (en) 1995-08-02 2000-02-22 Affymetrix, Inc. Arrays of nucleic acid probes and methods of using the same for detecting cystic fibrosis
US5650274A (en) * 1993-06-25 1997-07-22 Hitachi, Ltd. DNA analyzing method
DE69431719T2 (en) 1993-06-25 2003-09-18 Affymetrix Inc N D Ges D Staat HYBRIDIZATION AND SEQUENCING OF NUCLEIC ACIDS
US5837832A (en) * 1993-06-25 1998-11-17 Affymetrix, Inc. Arrays of nucleic acid probes on biological chips
US7115364B1 (en) 1993-10-26 2006-10-03 Affymetrix, Inc. Arrays of nucleic acid probes on biological chips
US5861242A (en) * 1993-06-25 1999-01-19 Affymetrix, Inc. Array of nucleic acid probes on biological chips for diagnosis of HIV and methods of using the same
WO1995003401A1 (en) 1993-07-23 1995-02-02 Hyseq, Inc. Method for screening unknown organisms
CN1525171A (en) * 1993-10-01 2004-09-01 ŦԼ�и��ױ��Ǵ�ѧ���� Complex combinatorial chemical libraries encoded with tags
JP3457322B2 (en) * 1993-10-21 2003-10-14 アボツト・ラボラトリーズ Apparatus and method for detecting target ligand
US6309823B1 (en) 1993-10-26 2001-10-30 Affymetrix, Inc. Arrays of nucleic acid probes for analyzing biotransformation genes and methods of using the same
US6045996A (en) * 1993-10-26 2000-04-04 Affymetrix, Inc. Hybridization assays on oligonucleotide arrays
US20060229824A1 (en) 1993-10-26 2006-10-12 Affymetrix, Inc. Arrays of nucleic acid probes for analyzing biotransformation genes
ZA95260B (en) * 1994-01-13 1995-09-28 Univ Columbia Synthetic receptors libraries and uses thereof
US6015880A (en) * 1994-03-16 2000-01-18 California Institute Of Technology Method and substrate for performing multiple sequential reactions on a matrix
US6936477B2 (en) 1994-04-13 2005-08-30 The Trustees Of Columbia University In The City Of New York Complex combinatorial chemical libraries encoded with tags
WO1995030774A1 (en) * 1994-05-05 1995-11-16 Beckman Instruments, Inc. Oligonucleotide repeat arrays
US5571639A (en) * 1994-05-24 1996-11-05 Affymax Technologies N.V. Computer-aided engineering system for design of sequence arrays and lithographic masks
US6287850B1 (en) 1995-06-07 2001-09-11 Affymetrix, Inc. Bioarray chip reaction apparatus and its manufacture
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
US6558633B1 (en) * 1994-09-21 2003-05-06 Isis Pharmaceuticals, Inc. Chemical reaction apparatus and methods
US5599668A (en) * 1994-09-22 1997-02-04 Abbott Laboratories Light scattering optical waveguide method for detecting specific binding events
US6280935B1 (en) 1994-10-13 2001-08-28 Lynx Therapeutics, Inc. Method of detecting the presence or absence of a plurality of target sequences using oligonucleotide tags
US5846719A (en) 1994-10-13 1998-12-08 Lynx Therapeutics, Inc. Oligonucleotide tags for sorting and identification
USRE43097E1 (en) 1994-10-13 2012-01-10 Illumina, Inc. Massively parallel signature sequencing by ligation of encoded adaptors
US5604097A (en) * 1994-10-13 1997-02-18 Spectragen, Inc. Methods for sorting polynucleotides using oligonucleotide tags
US5695934A (en) * 1994-10-13 1997-12-09 Lynx Therapeutics, Inc. Massively parallel sequencing of sorted polynucleotides
US5795716A (en) 1994-10-21 1998-08-18 Chee; Mark S. Computer-aided visualization and analysis system for sequence evaluation
US6600996B2 (en) 1994-10-21 2003-07-29 Affymetrix, Inc. Computer-aided techniques for analyzing biological sequences
US6974666B1 (en) * 1994-10-21 2005-12-13 Appymetric, Inc. Methods of enzymatic discrimination enhancement and surface-bound double-stranded DNA
US5747248A (en) * 1994-12-05 1998-05-05 Chiron Corporation Discontinuous probe design using hybritope mapping
US5830645A (en) 1994-12-09 1998-11-03 The Regents Of The University Of California Comparative fluorescence hybridization to nucleic acid arrays
US5908926A (en) * 1995-03-16 1999-06-01 Duke University 5'to 3' nucleic acid synthesis using 3'-photoremovable protecting group
US6312894B1 (en) 1995-04-03 2001-11-06 Epoch Pharmaceuticals, Inc. Hybridization and mismatch discrimination using oligonucleotides conjugated to minor groove binders
US5801155A (en) 1995-04-03 1998-09-01 Epoch Pharmaceuticals, Inc. Covalently linked oligonucleotide minor grove binder conjugates
US5753439A (en) * 1995-05-19 1998-05-19 Trustees Of Boston University Nucleic acid detection methods
US5614386A (en) * 1995-06-23 1997-03-25 Baylor College Of Medicine Alternative dye-labeled primers for automated DNA sequencing
US5968740A (en) * 1995-07-24 1999-10-19 Affymetrix, Inc. Method of Identifying a Base in a Nucleic Acid
US5633724A (en) * 1995-08-29 1997-05-27 Hewlett-Packard Company Evanescent scanning of biochemical array
US5733729A (en) 1995-09-14 1998-03-31 Affymetrix, Inc. Computer-aided probability base calling for arrays of nucleic acid probes on chips
US6300063B1 (en) 1995-11-29 2001-10-09 Affymetrix, Inc. Polymorphism detection
US6953663B1 (en) 1995-11-29 2005-10-11 Affymetrix, Inc. Polymorphism detection
US6864059B2 (en) 1996-01-23 2005-03-08 Affymetrix, Inc. Biotin containing C-glycoside nucleic acid labeling compounds
US20010018514A1 (en) 1998-07-31 2001-08-30 Mcgall Glenn H. Nucleic acid labeling compounds
US7282327B2 (en) 1996-01-23 2007-10-16 Affymetrix, Inc. Nucleic acid labeling compounds
US7291463B2 (en) 1996-01-23 2007-11-06 Affymetrix, Inc. Nucleic acid labeling compounds
US6965020B2 (en) 1996-01-23 2005-11-15 Affymetrix, Inc. Nucleic acid labeling compounds
US7423143B2 (en) 1996-01-23 2008-09-09 Affymetrix. Inc. Nucleic acid labeling compounds
US6924094B1 (en) 1996-02-08 2005-08-02 Affymetrix, Inc. Chip-based species identification and phenotypic characterization of microorganisms
AU2189397A (en) * 1996-02-08 1997-08-28 Affymetrix, Inc. Chip-based speciation and phenotypic characterization of microorganisms
EP1728875A3 (en) * 1996-02-08 2007-05-09 Affymetrix, Inc. Chip-based speciation and phenotypic characterization of microorganisms
EP1958955B1 (en) 1996-02-09 2013-09-04 Cornell Research Foundation, Inc. Spatially addressable oligonucleotide arrays and method of making the same
US6852487B1 (en) 1996-02-09 2005-02-08 Cornell Research Foundation, Inc. Detection of nucleic acid sequence differences using the ligase detection reaction with addressable arrays
US6361937B1 (en) 1996-03-19 2002-03-26 Affymetrix, Incorporated Computer-aided nucleic acid sequencing
US5958342A (en) * 1996-05-17 1999-09-28 Incyte Pharmaceuticals, Inc. Jet droplet device
EP2369007B1 (en) 1996-05-29 2015-07-29 Cornell Research Foundation, Inc. Detection of nucleic acid sequence differences using coupled ligase detection and polymerase chain reactions
US6391550B1 (en) 1996-09-19 2002-05-21 Affymetrix, Inc. Identification of molecular sequence signatures and methods involving the same
FR2754344B1 (en) 1996-10-04 1998-11-20 Bio Merieux METHOD AND DEVICE FOR THE COMPLEX TREATMENT OF A LIQUID MEDIUM
US6303301B1 (en) 1997-01-13 2001-10-16 Affymetrix, Inc. Expression monitoring for gene function identification
US6297006B1 (en) 1997-01-16 2001-10-02 Hyseq, Inc. Methods for sequencing repetitive sequences and for determining the order of sequence subfragments
US6309824B1 (en) 1997-01-16 2001-10-30 Hyseq, Inc. Methods for analyzing a target nucleic acid using immobilized heterogeneous mixtures of oligonucleotide probes
US20020042048A1 (en) 1997-01-16 2002-04-11 Radoje Drmanac Methods and compositions for detection or quantification of nucleic acid species
US5812272A (en) * 1997-01-30 1998-09-22 Hewlett-Packard Company Apparatus and method with tiled light source array for integrated assay sensing
US5837475A (en) * 1997-01-30 1998-11-17 Hewlett-Packard Co. Apparatus and method for scanning a chemical array
JP3935509B2 (en) 1997-02-12 2007-06-27 ワイ. チャン,ユージーン Methods and products for polymer analysis
IL131978A0 (en) 1997-03-20 2001-03-19 Univ Washington Solvent for biopolymer synthesis solvent microdots and methods of use
US6419883B1 (en) 1998-01-16 2002-07-16 University Of Washington Chemical synthesis using solvent microdroplets
US6028189A (en) * 1997-03-20 2000-02-22 University Of Washington Solvent for oligonucleotide synthesis and methods of use
EP1007737A4 (en) 1997-07-25 2002-07-03 Affymetrix Inc System for providing a polymorphism database
US6420108B2 (en) 1998-02-09 2002-07-16 Affymetrix, Inc. Computer-aided display for comparative gene expression
US6826296B2 (en) 1997-07-25 2004-11-30 Affymetrix, Inc. Method and system for providing a probe array chip design database
US7068830B2 (en) 1997-07-25 2006-06-27 Affymetrix, Inc. Method and system for providing a probe array chip design database
EP1003914A1 (en) * 1997-08-05 2000-05-31 Ambion, Inc. Methods and compositions for stripping nucleic acids
US6365731B1 (en) 1997-08-06 2002-04-02 Ambion, Inc. Stripping nucleic acids with iodine and sodium thiosulfate
JP2001514906A (en) * 1997-08-15 2001-09-18 ハイセック,インコーポレーテッド Methods and compositions for detecting or quantifying nucleic acid species
US6607878B2 (en) 1997-10-06 2003-08-19 Stratagene Collections of uniquely tagged molecules
US7351578B2 (en) 1999-12-10 2008-04-01 Invitrogen Corp. Use of multiple recombination sites with unique specificity in recombinational cloning
WO1999037816A1 (en) * 1998-01-26 1999-07-29 Genzyme Corporation Methods for identifying therapeutic targets
CA2318175A1 (en) 1998-02-04 1999-08-12 Invitrogen Corporation Microarrays and uses therefor
US6187535B1 (en) 1998-02-18 2001-02-13 Institut Pasteur Fast and exhaustive method for selecting a prey polypeptide interacting with a bait polypeptide of interest: application to the construction of maps of interactors polypeptides
US6127121A (en) * 1998-04-03 2000-10-03 Epoch Pharmaceuticals, Inc. Oligonucleotides containing pyrazolo[3,4-D]pyrimidines for hybridization and mismatch discrimination
US7045610B2 (en) 1998-04-03 2006-05-16 Epoch Biosciences, Inc. Modified oligonucleotides for mismatch discrimination
US7715989B2 (en) 1998-04-03 2010-05-11 Elitech Holding B.V. Systems and methods for predicting oligonucleotide melting temperature (TmS)
AU3463699A (en) * 1998-04-03 1999-10-25 Phylos, Inc. Addressable protein arrays
US6949367B1 (en) 1998-04-03 2005-09-27 Epoch Pharmaceuticals, Inc. Modified oligonucleotides for mismatch discrimination
US6683173B2 (en) 1998-04-03 2004-01-27 Epoch Biosciences, Inc. Tm leveling methods
US6355419B1 (en) 1998-04-27 2002-03-12 Hyseq, Inc. Preparation of pools of nucleic acids based on representation in a sample
US6780591B2 (en) 1998-05-01 2004-08-24 Arizona Board Of Regents Method of determining the nucleotide sequence of oligonucleotides and DNA molecules
US7875440B2 (en) 1998-05-01 2011-01-25 Arizona Board Of Regents Method of determining the nucleotide sequence of oligonucleotides and DNA molecules
US6699659B2 (en) 1998-06-01 2004-03-02 Affymetrix, Inc. Products and methods for analyzing nucleic acids including identification of substitutions, insertions and deletions
US6383744B1 (en) * 1998-07-10 2002-05-07 Incyte Genomics, Inc. Human checkpoint kinase
US6576478B1 (en) * 1998-07-14 2003-06-10 Zyomyx, Inc. Microdevices for high-throughput screening of biomolecules
US20020119579A1 (en) * 1998-07-14 2002-08-29 Peter Wagner Arrays devices and methods of use thereof
US6780582B1 (en) * 1998-07-14 2004-08-24 Zyomyx, Inc. Arrays of protein-capture agents and methods of use thereof
US6406921B1 (en) * 1998-07-14 2002-06-18 Zyomyx, Incorporated Protein arrays for high-throughput screening
US20030138973A1 (en) * 1998-07-14 2003-07-24 Peter Wagner Microdevices for screening biomolecules
US6185561B1 (en) 1998-09-17 2001-02-06 Affymetrix, Inc. Method and apparatus for providing and expression data mining database
US7071324B2 (en) 1998-10-13 2006-07-04 Brown University Research Foundation Systems and methods for sequencing by hybridization
US7034143B1 (en) * 1998-10-13 2006-04-25 Brown University Research Foundation Systems and methods for sequencing by hybridization
US6489096B1 (en) 1998-10-15 2002-12-03 Princeton University Quantitative analysis of hybridization patterns and intensities in oligonucleotide arrays
US6861218B2 (en) 1998-11-03 2005-03-01 Metasystems Hard And Software Gmbh Method for the targeted application of reagents onto immobilized biological material
EP0998974B1 (en) * 1998-11-03 2003-06-25 MetaSystems Hard & Software GmbH Method for the targetted application of reagents to immobilised biological material
WO2000034525A1 (en) 1998-12-09 2000-06-15 Vistagen, Inc. Toxicity typing using embryoid bodies
EP1196627B1 (en) * 1999-03-10 2005-12-07 Cold Spring Harbor Laboratory Gene chip technology for determining memory genes
US7005256B1 (en) 1999-03-10 2006-02-28 Cold Spring Harbor Laboratory Gene chip technology for determining memory genes
US6506594B1 (en) 1999-03-19 2003-01-14 Cornell Res Foundation Inc Detection of nucleic acid sequence differences using the ligase detection reaction with addressable arrays
US7014994B1 (en) 1999-03-19 2006-03-21 Cornell Research Foundation,Inc. Coupled polymerase chain reaction-restriction-endonuclease digestion-ligase detection reaction process
US20030207295A1 (en) * 1999-04-20 2003-11-06 Kevin Gunderson Detection of nucleic acid reactions on bead arrays
US20060275782A1 (en) * 1999-04-20 2006-12-07 Illumina, Inc. Detection of nucleic acid reactions on bead arrays
AU5269600A (en) * 1999-05-13 2000-12-05 Oligos Etc. Inc. Arrays with modified oligonucleotide and polynucleotide compositions
US7056661B2 (en) 1999-05-19 2006-06-06 Cornell Research Foundation, Inc. Method for sequencing nucleic acid molecules
DE60000583T3 (en) 1999-05-19 2009-04-30 Eppendorf Array Technologies METHOD FOR IDENTIFYING AND / OR QUANTIFYING A TARGET LINK
US6818395B1 (en) 1999-06-28 2004-11-16 California Institute Of Technology Methods and apparatus for analyzing polynucleotide sequences
US6346423B1 (en) 1999-07-16 2002-02-12 Agilent Technologies, Inc. Methods and compositions for producing biopolymeric arrays
US6339147B1 (en) 1999-07-29 2002-01-15 Epoch Biosciences, Inc. Attachment of oligonucleotides to solid supports through Schiff base type linkages for capture and detection of nucleic acids
CA2382436C (en) 1999-08-30 2011-05-17 Illumina, Inc. Methods for improving signal detection from an array
US6607885B1 (en) 1999-10-15 2003-08-19 E. I. Du Pont De Nemours And Company Method for high-density microarray medicated gene expression profiling
US6511806B1 (en) * 1999-11-03 2003-01-28 Oncotech, Inc. Methods for cancer prognosis and diagnosis
US6660845B1 (en) 1999-11-23 2003-12-09 Epoch Biosciences, Inc. Non-aggregating, non-quenching oligomers comprising nucleotide analogues; methods of synthesis and use thereof
US6727356B1 (en) 1999-12-08 2004-04-27 Epoch Pharmaceuticals, Inc. Fluorescent quenching detection reagents and methods
US20040081959A9 (en) 1999-12-08 2004-04-29 Epoch Biosciences, Inc. Fluorescent quenching detection reagents and methods
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
US7955794B2 (en) * 2000-09-21 2011-06-07 Illumina, Inc. Multiplex nucleic acid reactions
US7582420B2 (en) 2001-07-12 2009-09-01 Illumina, Inc. Multiplex nucleic acid reactions
US6770441B2 (en) * 2000-02-10 2004-08-03 Illumina, Inc. Array compositions and methods of making same
EP1944310A3 (en) 2000-03-01 2008-08-06 Epoch Biosciences, Inc. Modified oligonucleotides for mismatch discrimination
ES2391213T3 (en) 2000-03-01 2012-11-22 Epoch Biosciences, Inc. Modified oligonucleotides for mismatch discrimination
WO2001079548A2 (en) 2000-04-14 2001-10-25 Cornell Research Foundation, Inc. Method of designing addressable array for detection of nucleic acid sequence differences using ligase detection reaction
AU2001259512B2 (en) 2000-05-04 2007-03-01 Yale University High density protein arrays for screening of protein activity
KR100865664B1 (en) 2000-06-14 2008-10-29 비스타겐 인코포레이티드 Toxicity typing using liver stem cells
US6954253B2 (en) * 2000-07-25 2005-10-11 Scientific Solutions, Inc. Optical multiplexer and cross-switch using etched liquid crystal fabry-perot etalons
DE10036174B4 (en) 2000-07-25 2006-12-07 Axaron Bioscience Ag Method and apparatus for analyzing chemical or biological samples
US6984522B2 (en) 2000-08-03 2006-01-10 Regents Of The University Of Michigan Isolation and use of solid tumor stem cells
US7198924B2 (en) 2000-12-11 2007-04-03 Invitrogen Corporation Methods and compositions for synthesis of nucleic acid molecules using multiple recognition sites
US6629038B1 (en) * 2000-09-05 2003-09-30 Yeda Research And Development Co. Ltd. Method and system for identifying commercially distributed organisms
WO2002044425A2 (en) 2000-12-01 2002-06-06 Visigen Biotechnologies, Inc. Enzymatic nucleic acid synthesis: compositions and methods for altering monomer incorporation fidelity
DE10105339B4 (en) * 2001-02-05 2004-05-13 november Aktiengesellschaft Gesellschaft für Molekulare Medizin Counterfeit-proof marking method, counterfeit-proof marking and kit
EP1360336A2 (en) * 2001-02-15 2003-11-12 Bayer Corporation Innate immunity markers for rapid diagnosis of infectious diseases
EP1385829A4 (en) 2001-03-12 2005-10-19 Affymetrix Inc Nucleic acid labeling compounds
US7297518B2 (en) * 2001-03-12 2007-11-20 California Institute Of Technology Methods and apparatus for analyzing polynucleotide sequences by asynchronous base extension
US7138506B2 (en) 2001-05-09 2006-11-21 Genetic Id, Na, Inc. Universal microarray system
US7196183B2 (en) 2001-08-31 2007-03-27 Innogenetics N.V. Hepatitis C virus genotype, and its use as prophylactic, therapeutic and diagnostic agent
WO2004013290A2 (en) * 2002-08-05 2004-02-12 Invitrogen Corporation Compositions and methods for molecular biology
US20040053315A1 (en) * 2002-08-12 2004-03-18 Caliper Technologies Corp. Methods and systems for monitoring molecular interactions
US20050233473A1 (en) * 2002-08-16 2005-10-20 Zyomyx, Inc. Methods and reagents for surface functionalization
JP2005536769A (en) * 2002-08-20 2005-12-02 シヴェラ コーポレイション Optical identification elements based on diffraction gratings
US7872804B2 (en) 2002-08-20 2011-01-18 Illumina, Inc. Encoded particle having a grating with variations in the refractive index
US7441703B2 (en) * 2002-08-20 2008-10-28 Illumina, Inc. Optical reader for diffraction grating-based encoded optical identification elements
US7901630B2 (en) 2002-08-20 2011-03-08 Illumina, Inc. Diffraction grating-based encoded microparticle assay stick
US7923260B2 (en) 2002-08-20 2011-04-12 Illumina, Inc. Method of reading encoded particles
US7900836B2 (en) 2002-08-20 2011-03-08 Illumina, Inc. Optical reader system for substrates having an optically readable code
US7508608B2 (en) 2004-11-17 2009-03-24 Illumina, Inc. Lithographically fabricated holographic optical identification element
US7164533B2 (en) 2003-01-22 2007-01-16 Cyvera Corporation Hybrid random bead/chip based microarray
CA2499046A1 (en) * 2002-09-12 2004-03-25 Cyvera Corporation Diffraction grating-based encoded micro-particles for multiplexed experiments
US7092160B2 (en) 2002-09-12 2006-08-15 Illumina, Inc. Method of manufacturing of diffraction grating-based optical identification element
US20100255603A9 (en) 2002-09-12 2010-10-07 Putnam Martin A Method and apparatus for aligning microbeads in order to interrogate the same
CA2499037A1 (en) * 2002-09-12 2004-03-25 Cyvera Corporation Method and apparatus for labelling using diffraction grating-based encoded optical identification elements
US7595883B1 (en) 2002-09-16 2009-09-29 The Board Of Trustees Of The Leland Stanford Junior University Biological analysis arrangement and approach therefor
US7498176B2 (en) * 2002-09-27 2009-03-03 Roche Nimblegen, Inc. Microarray with hydrophobic barriers
US20040110212A1 (en) * 2002-09-30 2004-06-10 Mccormick Mark Microarrays with visual alignment marks
CA2500783C (en) * 2002-10-01 2012-07-17 Nimblegen Systems, Inc. Microarrays having multiple oligonucleotides in single array features
US20040259105A1 (en) * 2002-10-03 2004-12-23 Jian-Bing Fan Multiplex nucleic acid analysis using archived or fixed samples
US9453251B2 (en) 2002-10-08 2016-09-27 Pfenex Inc. Expression of mammalian proteins in Pseudomonas fluorescens
AU2002952696A0 (en) 2002-11-14 2002-11-28 Genomics Research Partners Pty Ltd Status determination
WO2004046387A1 (en) * 2002-11-15 2004-06-03 Sangamo Biosciences, Inc. Methods and compositions for analysis of regulatory sequences
US7250289B2 (en) 2002-11-20 2007-07-31 Affymetrix, Inc. Methods of genetic analysis of mouse
US7314750B2 (en) 2002-11-20 2008-01-01 Affymetrix, Inc. Addressable oligonucleotide array of the rat genome
US7541152B2 (en) 2002-12-24 2009-06-02 Agilent Technologies, Inc. Integrated light source for diagnostic arrays
CA2458085A1 (en) 2003-03-21 2004-09-21 F. Hoffmann-La Roche Ag Transcriptional activity assay
US7833714B1 (en) * 2003-04-11 2010-11-16 Affymetrix, Inc. Combinatorial affinity selection
US20040248323A1 (en) * 2003-06-09 2004-12-09 Protometrix, Inc. Methods for conducting assays for enzyme activity on protein microarrays
WO2005003301A2 (en) * 2003-06-17 2005-01-13 Signal Pharmaceuticals, Inc. Methods, compositions, and kits for predicting the effect of compounds on hot flash symptoms
US7642064B2 (en) 2003-06-24 2010-01-05 Ventana Medical Systems, Inc. Enzyme-catalyzed metal deposition for the enhanced detection of analytes of interest
ES2596753T3 (en) 2003-08-29 2017-01-11 Fujirebio Europe N.V. New clade of the hepatitis C virus and prototype sequences thereof
US8131475B2 (en) 2003-09-03 2012-03-06 The United States Of America As Represented By The Secretary, Department Of Health And Human Services Methods for identifying, diagnosing, and predicting survival of lymphomas
EP1664762A4 (en) 2003-09-03 2008-08-13 Us Gov Health & Human Serv Methods for identifying, diagnosing, and predicting survival of lymphomas
US7365179B2 (en) * 2003-09-09 2008-04-29 Compass Genetics, Llc Multiplexed analytical platform
US20050100939A1 (en) * 2003-09-18 2005-05-12 Eugeni Namsaraev System and methods for enhancing signal-to-noise ratios of microarray-based measurements
WO2005034070A2 (en) * 2003-10-01 2005-04-14 Board Of Regents The University Of Texas System Compositions, methods and systems for making and using electronic paper
US20080297878A1 (en) * 2003-10-01 2008-12-04 Board Of Regents, The University Of Texas System Compositions, methods and systems for making and using electronic paper
US20050227251A1 (en) 2003-10-23 2005-10-13 Robert Darnell Method of purifying RNA binding protein-RNA complexes
EP1678329A4 (en) * 2003-10-30 2008-07-02 Tufts New England Medical Ct Prenatal diagnosis using cell-free fetal dna in amniotic fluid
US7169560B2 (en) 2003-11-12 2007-01-30 Helicos Biosciences Corporation Short cycle methods for sequencing polynucleotides
US7439341B2 (en) 2003-11-14 2008-10-21 Integrated Dna Technologies, Inc. Fluorescence quenching azo dyes, their methods of preparation and use
EP2287341B1 (en) 2003-12-01 2013-02-13 Life Technologies Corporation Nucleic acid molecules containing recombination sites and methods of using the same
WO2005070020A2 (en) 2004-01-23 2005-08-04 The Regents Of The University Of Colorado Gefitinib sensitivity-related gene expression and products and methods related thereto
AU2005214329A1 (en) * 2004-02-12 2005-09-01 Population Genetics Technologies Ltd Genetic analysis by sequence-specific sorting
ATE463584T1 (en) 2004-02-19 2010-04-15 Helicos Biosciences Corp METHOD FOR ANALYZING POLYNUCLEOTIDE SEQUENCES
US7433123B2 (en) 2004-02-19 2008-10-07 Illumina, Inc. Optical identification element having non-waveguide photosensitive substrate with diffraction grating therein
WO2005111242A2 (en) * 2004-05-10 2005-11-24 Parallele Bioscience, Inc. Digital profiling of polynucleotide populations
AU2005249492B2 (en) 2004-05-27 2011-09-22 The Regents Of The University Of Colorado Methods for prediction of clinical outcome to epidermal growth factor receptor inhibitors by cancer patients
WO2006020363A2 (en) 2004-07-21 2006-02-23 Illumina, Inc. Method and apparatus for drug product tracking using encoded optical identification elements
JP2008507294A (en) 2004-07-26 2008-03-13 ダウ グローバル テクノロジーズ インコーポレイティド Method for improved protein expression by strain genetic manipulation
US20060019304A1 (en) * 2004-07-26 2006-01-26 Paul Hardenbol Simultaneous analysis of multiple genomes
DE102004038163A1 (en) * 2004-08-06 2006-03-16 Diacdem Chip Technologies Gmbh Fluorescence-based assays for the rapid, quantitative analysis of biomolecules (proteins and nucleic acids) by enrichment on cells or beads
US7170050B2 (en) 2004-09-17 2007-01-30 Pacific Biosciences Of California, Inc. Apparatus and methods for optical analysis of molecules
ATE428421T1 (en) 2004-09-17 2009-05-15 Eisai R&D Man Co Ltd MEDICAL COMPOSITION WITH IMPROVED STABILITY AND REDUCED GELING PROPERTIES
US7476503B2 (en) 2004-09-17 2009-01-13 Pacific Biosciences Of California, Inc. Apparatus and method for performing nucleic acid analysis
EP1647600A3 (en) 2004-09-17 2006-06-28 Affymetrix, Inc. (A US Entity) Methods for identifying biological samples by addition of nucleic acid bar-code tags
CA2587674A1 (en) 2004-11-16 2006-05-26 Illumina, Inc. Method and apparatus for reading coded microbeads
US20060183893A1 (en) * 2005-01-25 2006-08-17 North Don A Nucleic acids for apoptosis of cancer cells
ATE429495T1 (en) * 2005-01-25 2009-05-15 Sky Genetics Inc NUCLEIC ACIDS FOR THE APOPTOSIS OF CANCER CELLS
US7407757B2 (en) * 2005-02-10 2008-08-05 Population Genetics Technologies Genetic analysis by sequence-specific sorting
US8178291B2 (en) 2005-02-18 2012-05-15 Monogram Biosciences, Inc. Methods and compositions for determining hypersusceptibility of HIV-1 to non-nucleoside reverse transcriptase inhibitors
EP2522757A1 (en) 2005-02-18 2012-11-14 Monogram BioSciences, Inc. Methods and compositions for determining anti-HIV drug susceptibility and replication capacity of HIV
US20060211030A1 (en) * 2005-03-16 2006-09-21 Sydney Brenner Methods and compositions for assay readouts on multiple analytical platforms
US20070020650A1 (en) * 2005-04-01 2007-01-25 Avak Kahvejian Methods for detecting proteins
AU2006251637B2 (en) 2005-05-20 2012-06-14 Integrated Dna Technologies, Inc. Compounds and methods for labeling oligonucleotides
WO2006130477A2 (en) * 2005-05-27 2006-12-07 Monogram Biosciences, Inc. Methods and compositions for determining resistance of hiv-1 to protease inhibitors
US9506121B2 (en) 2005-06-06 2016-11-29 Monogram Biosciences, Inc. Methods for determining resistance or susceptibility to HIV entry inhibitors
US8071284B2 (en) * 2005-06-06 2011-12-06 Monogram Biosciences, Inc. Methods and compositions for determining altered susceptibility of HIV-1 to anti-HIV drugs
EP1925676A4 (en) 2005-08-02 2010-11-10 Eisai R&D Man Co Ltd Method for assay on the effect of vascularization inhibitor
CN101356285A (en) 2005-08-16 2009-01-28 梅洛根有限责任公司 Methods for identification of merle gene
US7666593B2 (en) 2005-08-26 2010-02-23 Helicos Biosciences Corporation Single molecule sequencing of captured nucleic acids
CN102716490A (en) * 2005-09-01 2012-10-10 卫材R&D管理有限公司 Method for preparation of pharmaceutical composition having improved disintegradability
WO2007081410A2 (en) * 2005-09-13 2007-07-19 True Materials Incorporated Encoded microparticles
US8178278B2 (en) * 2005-09-13 2012-05-15 Affymetrix, Inc. Miniaturized microparticles
CA2624896C (en) 2005-10-07 2017-11-07 Callida Genomics, Inc. Self-assembled single molecule arrays and uses thereof
CA2627598C (en) 2005-11-07 2013-06-25 Eisai R & D Management Co., Ltd. Use of combination of anti-angiogenic substance and c-kit kinase inhibitor
US7889347B2 (en) * 2005-11-21 2011-02-15 Plexera Llc Surface plasmon resonance spectrometer with an actuator driven angle scanning mechanism
US7623624B2 (en) * 2005-11-22 2009-11-24 Illumina, Inc. Method and apparatus for labeling using optical identification elements characterized by X-ray diffraction
US7463358B2 (en) 2005-12-06 2008-12-09 Lumera Corporation Highly stable surface plasmon resonance plates, microarrays, and methods
US20100009373A1 (en) * 2005-12-23 2010-01-14 Perkinelmer Health Sciences, Inc. Methods and compositions relating to multiplex genomic gain and loss assays
US7932037B2 (en) 2007-12-05 2011-04-26 Perkinelmer Health Sciences, Inc. DNA assays using amplicon probes on encoded particles
US20090104613A1 (en) * 2005-12-23 2009-04-23 Perkinelmer Las, Inc. Methods and compositions relating to multiplexed genomic gain and loss assays
US7537897B2 (en) * 2006-01-23 2009-05-26 Population Genetics Technologies, Ltd. Molecular counting
US9445025B2 (en) 2006-01-27 2016-09-13 Affymetrix, Inc. System, method, and product for imaging probe arrays with small feature sizes
WO2007121054A2 (en) * 2006-03-27 2007-10-25 Sky Genetics, Inc. Nucleic acids for apoptosis of cancer cells
US7285388B1 (en) 2006-04-07 2007-10-23 Merlogen, Llc Methods for identification of alport syndrome
US7888044B2 (en) 2006-04-10 2011-02-15 Trustees Of Boston University Melanoma biomarker and methods of uses
US7830575B2 (en) 2006-04-10 2010-11-09 Illumina, Inc. Optical scanner with improved scan time
EP2036557B1 (en) * 2006-05-18 2015-10-21 Eisai R&D Management Co., Ltd. Antitumor agent for thyroid cancer
CN106566855B (en) * 2006-07-31 2021-11-09 苏州新海生物科技股份有限公司 Nucleic acid amplification using reversibly modified oligonucleotides
EP2065372B1 (en) * 2006-08-28 2012-11-28 Eisai R&D Management Co., Ltd. Antitumor agent for undifferentiated gastric cancer
JP5319306B2 (en) * 2007-01-29 2013-10-16 エーザイ・アール・アンド・ディー・マネジメント株式会社 Composition for treatment of undifferentiated gastric cancer
WO2008106115A2 (en) 2007-02-26 2008-09-04 Monogram Biosciences, Inc. Compositions and methods for determining whether a subject would benefit from co-receptor inhibitor therapy
US9580719B2 (en) 2007-04-27 2017-02-28 Pfenex, Inc. Method for rapidly screening microbial hosts to identify certain strains with improved yield and/or quality in the expression of heterologous proteins
CA2964910C (en) 2007-04-27 2018-01-09 Pfenex Inc. Method for rapidly screening microbial hosts to identify certain strains with improved yield and/or quality in the expression of heterologous proteins
US8454895B2 (en) * 2007-05-03 2013-06-04 Nanyang Technological University Online contaminant detection and removal system
US20100303813A1 (en) 2007-06-08 2010-12-02 Biogen Idec Ma Inc. Biomarkers for predicting anti-tnf responsiveness or non-responsiveness
CA2692171C (en) 2007-06-22 2019-10-22 Randolph Watnick Methods and uses thereof of prosaposin
US20090060786A1 (en) * 2007-08-29 2009-03-05 Gibum Kim Microfluidic apparatus for wide area microarrays
CA2704000C (en) * 2007-11-09 2016-12-13 Eisai R&D Management Co., Ltd. Combination of anti-angiogenic substance and anti-tumor platinum complex
WO2009099967A1 (en) * 2008-02-01 2009-08-13 Life Technologies Corporation System and method for selecting samples from an antibody analysis for display of complete specificity analysis data
JP2011510675A (en) * 2008-02-07 2011-04-07 フォレンシック サイエンシーズ サーヴィス リミテッド Analytical and analytical improvements
JP2011515102A (en) 2008-03-28 2011-05-19 パシフィック バイオサイエンシーズ オブ カリフォルニア, インコーポレイテッド Compositions and methods for nucleic acid sequencing
WO2009137369A1 (en) * 2008-05-03 2009-11-12 Tufts Medical Center, Inc. Neonatal salivary genomics
JP2009268665A (en) * 2008-05-07 2009-11-19 Canon Inc Inhalation device
JP5892791B2 (en) 2008-05-14 2016-03-23 ミレニアム ファーマシューティカルズ, インコーポレイテッドMillennium Pharmaceuticals, Inc. Methods and kits for monitoring the effects of immunomodulators on adaptive immunity
TWI460602B (en) * 2008-05-16 2014-11-11 Counsyl Inc Device for universal preconception screening
CA2726811C (en) 2008-06-06 2019-11-26 The United States Of America, As Represented By The Secretary, Dept. Of Health And Human Services Survival predictor for diffuse large b cell lymphoma
WO2010028366A2 (en) * 2008-09-05 2010-03-11 Life Technologies Corporation Methods and systems for nucleic acid sequencing validation, calibration and normalization
CN102308004A (en) 2008-10-30 2012-01-04 卡里斯生命科学卢森堡控股有限责任公司 Methods for assessing RNA patterns
EP2393596B1 (en) * 2009-02-09 2016-09-28 Whitespace Enterprise Corporation Microfluidic devices and methods of providing a storable sample
GB2472371B (en) * 2009-04-24 2011-10-26 Selectamark Security Systems Plc Synthetic nucleotide containing compositions for use in security marking of property and/or for marking a thief or attacker
EP2248914A1 (en) 2009-05-05 2010-11-10 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. The use of class IIB restriction endonucleases in 2nd generation sequencing applications
US20120316080A1 (en) 2009-10-19 2012-12-13 Stichting Het Nederlands Kanker Instiuut Differentiation between brca2-associated tumours and sporadic tumours via array comparative genomic hybridization
US20120322677A1 (en) 2009-10-19 2012-12-20 Stichting Het Nederlands Kanker Instituut Predicting benefit of anti-cancer therapy via array comparative genomic hybridization
EP2491140A1 (en) 2009-10-19 2012-08-29 Stichting Het Nederlands Kanker Instituut Predicting response to anti-cancer therapy via array comparative genomic hybridization
WO2011068569A1 (en) * 2009-12-04 2011-06-09 Biotium, Inc. Heterocycle-substituted xanthene dyes
US8835358B2 (en) 2009-12-15 2014-09-16 Cellular Research, Inc. Digital counting of individual molecules by stochastic attachment of diverse labels
US9315857B2 (en) 2009-12-15 2016-04-19 Cellular Research, Inc. Digital counting of individual molecules by stochastic attachment of diverse label-tags
EP3925670A1 (en) 2009-12-17 2021-12-22 Children's Medical Center, Corp. Saposin-a derived peptides and uses thereof
WO2011103467A2 (en) 2010-02-19 2011-08-25 Life Technologies Corporation Methods and systems for nucleic acid sequencing validation, calibration and normalization
EP2544688B1 (en) 2010-03-02 2016-09-07 President and Fellows of Harvard College Methods and compositions for treatment of angelman syndrome
US20120108799A1 (en) 2010-09-07 2012-05-03 Integrated Dna Technologies, Inc. Modifications for Antisense Compounds
EP2553123B1 (en) 2010-03-26 2016-08-24 Integrated DNA Technologies, Inc. Methods for enhancing nucleic acid hybridization
US9506057B2 (en) 2010-03-26 2016-11-29 Integrated Dna Technologies, Inc. Modifications for antisense compounds
KR101677790B1 (en) 2010-06-25 2016-11-18 에자이 알앤드디 매니지먼트 가부시키가이샤 Antitumor agent using compounds having kinase inhibitory effect in combination
US20130296183A1 (en) 2010-09-17 2013-11-07 President And Fellows Of Harvard College Functional genomics assay for characterizing pluripotent stem cell utility and safety
EP2619326A2 (en) 2010-09-20 2013-07-31 Stichting Het Nederlands Kanker Instituut Methods for predicting response to anti-cancer therapy in cancer patients
CN103154273A (en) 2010-09-21 2013-06-12 群体遗传学科技有限公司 Increasing confidence of allele calls with molecular counting
WO2012129547A1 (en) 2011-03-23 2012-09-27 Elitech Holding B.V. Functionalized 3-alkynyl pyrazolopyrimidine analogues as universal bases and methods of use
US8969003B2 (en) 2011-03-23 2015-03-03 Elitech Holding B.V. Functionalized 3-alkynyl pyrazolopyrimidine analogues as universal bases and methods of use
US8962650B2 (en) 2011-04-18 2015-02-24 Eisai R&D Management Co., Ltd. Therapeutic agent for tumor
ES2537189T3 (en) 2011-05-24 2015-06-03 Elitech Holding B.V. Detection of methicillin-resistant Staphylococci
ES2705950T3 (en) 2011-06-03 2019-03-27 Eisai R&D Man Co Ltd Biomarkers to predict and assess the responsiveness of subjects with thyroid and kidney cancer to lenvatinib compounds
CA2839896A1 (en) 2011-06-21 2012-12-27 Alnylam Pharmaceuticals, Inc. Assays and methods for determining activity of a therapeutic agent in a subject
CA2839451A1 (en) 2011-06-27 2013-01-03 The Jackson Laboratory Methods and compositions for treatment of cancer and autoimmune disease
WO2013055911A1 (en) 2011-10-14 2013-04-18 Dana-Farber Cancer Institute, Inc. Znf365/zfp365 biomarker predictive of anti-cancer response
JP6238900B2 (en) 2011-10-28 2017-11-29 ミレニアム ファーマシューティカルズ, インコーポレイテッドMillennium Pharmaceuticals, Inc. Biomarkers of response to NAE inhibitors
CN104126017A (en) 2011-11-11 2014-10-29 米伦纽姆医药公司 Biomarkers of response to proteasome inhibitors
JP6397765B2 (en) 2011-11-11 2018-09-26 ミレニアム ファーマシューティカルズ, インコーポレイテッドMillennium Pharmaceuticals, Inc. Biomarkers that respond to proteasome inhibitors
WO2013074676A2 (en) 2011-11-14 2013-05-23 The General Hospital Corporation Assays and methods for selecting a treatment regimen for a subject with depression
AU2012358269B2 (en) 2011-12-22 2017-11-02 Children's Medical Center Corporation Saposin-A derived peptides and uses thereof
CN104364392B (en) 2012-02-27 2018-05-25 赛卢拉研究公司 For the composition and kit of numerator counts
WO2013130512A2 (en) 2012-02-27 2013-09-06 The University Of North Carolina At Chapel Hill Methods and uses for molecular tags
EP2820155B1 (en) 2012-02-28 2017-07-26 Population Genetics Technologies Ltd. Method for attaching a counter sequence to a nucleic acid sample
WO2013149258A2 (en) 2012-03-30 2013-10-03 Charles Drew University of Medicine and Science Compositions and methods for treating or preventing metabolic syndrome disorders
US11913065B2 (en) 2012-09-04 2024-02-27 Guardent Health, Inc. Systems and methods to detect rare mutations and copy number variation
US20160040229A1 (en) 2013-08-16 2016-02-11 Guardant Health, Inc. Systems and methods to detect rare mutations and copy number variation
US10876152B2 (en) 2012-09-04 2020-12-29 Guardant Health, Inc. Systems and methods to detect rare mutations and copy number variation
IL305303A (en) 2012-09-04 2023-10-01 Guardant Health Inc Systems and methods to detect rare mutations and copy number variation
CA2886783A1 (en) 2012-10-01 2014-04-10 Millennium Pharmaceuticals, Inc. Biomarkers and methods to predict response to inhibitors and uses thereof
KR20150098605A (en) 2012-12-21 2015-08-28 에자이 알앤드디 매니지먼트 가부시키가이샤 Amorphous form of quinoline derivative, and method for producing same
EP2971086A1 (en) 2013-03-14 2016-01-20 ELITechGroup B.V. Functionalized 3-alkynyl pyrazolopyrimidine analogues as universal bases and methods of use
US9146248B2 (en) 2013-03-14 2015-09-29 Intelligent Bio-Systems, Inc. Apparatus and methods for purging flow cells in nucleic acid sequencing instruments
US20140274747A1 (en) * 2013-03-15 2014-09-18 Illumina, Inc. Super resolution imaging
US9591268B2 (en) 2013-03-15 2017-03-07 Qiagen Waltham, Inc. Flow cell alignment methods and systems
MX368099B (en) 2013-05-14 2019-09-19 Eisai R&D Man Co Ltd Biomarkers for predicting and assessing responsiveness of endometrial cancer subjects to lenvatinib compounds.
JP6697380B2 (en) 2013-06-10 2020-05-20 プレジデント・アンド・フェロウズ・オブ・ハーバード・カレッジ Early developmental genomic assay to characterize the utility and safety of pluripotent stem cells
EP3722442B1 (en) 2013-08-05 2023-04-05 Twist Bioscience Corporation De novo synthesized gene libraries
WO2015031691A1 (en) 2013-08-28 2015-03-05 Cellular Research, Inc. Massively parallel single cell analysis
US9501796B2 (en) * 2013-09-18 2016-11-22 Chicago Mercantile Exchange Inc. Dataset intersection determination
US9582877B2 (en) 2013-10-07 2017-02-28 Cellular Research, Inc. Methods and systems for digitally counting features on arrays
JP6657105B2 (en) 2013-11-06 2020-03-04 アメリカ合衆国 Methods for subtyping lymphoma types by expression profiling
AU2014369841B2 (en) 2013-12-28 2019-01-24 Guardant Health, Inc. Methods and systems for detecting genetic variants
WO2015164743A2 (en) 2014-04-24 2015-10-29 Dana-Farber Cancer Institute, Inc. Tumor suppressor and oncogene biomarkers predictive of anti-immune checkpoint inhibitor response
AU2015276899B2 (en) 2014-06-19 2021-08-12 Memorial Sloan-Kettering Cancer Center Biomarkers for response to EZH2 inhibitors
WO2016011383A1 (en) 2014-07-17 2016-01-21 The Trustees Of The University Of Pennsylvania Methods for using exosomes to monitor transplanted organ status
KR20230043234A (en) 2014-08-28 2023-03-30 에자이 알앤드디 매니지먼트 가부시키가이샤 High-purity quinoline derivative and method for manufacturing same
US20170248603A1 (en) 2014-10-06 2017-08-31 Dana-Farber Cancer Institute, Inc. Angiopoiten-2 biomarkers predictive of anti-immune checkpoint response
CN107106876B (en) 2014-10-09 2021-06-11 丹娜法伯癌症研究院 Multiple-variable IL-2 dosage regimen for treating immune disorders
PL3224376T5 (en) * 2014-11-28 2023-08-21 Uniqure Ip B.V. Dna impurities in a composition comprising a parvoviral virion
TW201702218A (en) 2014-12-12 2017-01-16 美國杰克森實驗室 Compositions and methods relating to the treatment of cancer, autoimmune disease, and neurodegenerative disease
WO2016123454A1 (en) 2015-01-29 2016-08-04 Board Of Trustees Of Miching State University Cryptic polypeptides and uses thereof
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
EP3766988B1 (en) 2015-02-19 2024-02-14 Becton, Dickinson and Company High-throughput single-cell analysis combining proteomic and genomic information
EP3263106B1 (en) 2015-02-25 2023-10-25 Eisai R&D Management Co., Ltd. Method for suppressing bitterness of quinoline derivative
EP3262192B1 (en) 2015-02-27 2020-09-16 Becton, Dickinson and Company Spatially addressable molecular barcoding
WO2016140717A1 (en) 2015-03-04 2016-09-09 Merck Sharp & Dohme Corp. Combination of a pd-1 antagonist and a vegfr/fgfr/ret tyrosine kinase inhibitor for treating cancer
CA2977532A1 (en) 2015-03-06 2016-09-15 Dana-Farber Cancer Institute, Inc. Pd-l2 biomarkers predictive of pd-1 pathway inhibitor responses in esophagogastric cancers
JP2018509915A (en) 2015-03-30 2018-04-12 セルラー リサーチ, インコーポレイテッド Methods and compositions for combinatorial bar coding
WO2016172377A1 (en) 2015-04-21 2016-10-27 Twist Bioscience Corporation Devices and methods for oligonucleic acid library synthesis
US11390914B2 (en) 2015-04-23 2022-07-19 Becton, Dickinson And Company Methods and compositions for whole transcriptome amplification
WO2016196229A1 (en) 2015-06-01 2016-12-08 Cellular Research, Inc. Methods for rna quantification
MX2017015896A (en) 2015-06-16 2018-08-09 Eisai R&D Man Co Ltd Anticancer agent.
EP3347465B1 (en) 2015-09-11 2019-06-26 Cellular Research, Inc. Methods and compositions for nucleic acid library normalization
KR20180050411A (en) 2015-09-18 2018-05-14 트위스트 바이오사이언스 코포레이션 Oligonucleotide mutant library and its synthesis
CN113604546A (en) 2015-09-22 2021-11-05 特韦斯特生物科学公司 Flexible substrates for nucleic acid synthesis
CN108603307A (en) 2015-12-01 2018-09-28 特韦斯特生物科学公司 functionalized surface and its preparation
SG11201805119QA (en) 2015-12-17 2018-07-30 Guardant Health Inc Methods to determine tumor gene copy number by analysis of cell-free dna
WO2017184861A1 (en) 2016-04-20 2017-10-26 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Evaluation of mantle cell lymphoma and methods related thereto
US10822643B2 (en) 2016-05-02 2020-11-03 Cellular Research, Inc. Accurate molecular barcoding
US10301677B2 (en) 2016-05-25 2019-05-28 Cellular Research, Inc. Normalization of nucleic acid libraries
JP7046007B2 (en) 2016-05-26 2022-04-01 ベクトン・ディキンソン・アンド・カンパニー How to adjust the molecular label count
US10202641B2 (en) 2016-05-31 2019-02-12 Cellular Research, Inc. Error correction in amplification of samples
US10640763B2 (en) 2016-05-31 2020-05-05 Cellular Research, Inc. Molecular indexing of internal sequences
GB2568444A (en) 2016-08-22 2019-05-15 Twist Bioscience Corp De novo synthesized nucleic acid libraries
AU2017332721B2 (en) 2016-09-20 2023-11-09 Sara BUHRLAGE Compositions and methods for identification, assessment, prevention, and treatment of AML using USP10 biomarkers and modulators
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
CN115060911A (en) 2016-09-26 2022-09-16 贝克顿迪金森公司 Measurement of protein expression using reagents with barcoded oligonucleotide sequences
US11608497B2 (en) 2016-11-08 2023-03-21 Becton, Dickinson And Company Methods for cell label classification
WO2018089378A1 (en) 2016-11-08 2018-05-17 Cellular Research, Inc. Methods for expression profile classification
WO2018112426A1 (en) 2016-12-16 2018-06-21 Twist Bioscience Corporation Variant libraries of the immunological synapse and synthesis thereof
CN110573253B (en) 2017-01-13 2021-11-02 赛卢拉研究公司 Hydrophilic coating for fluid channels
CN110382708A (en) 2017-02-01 2019-10-25 赛卢拉研究公司 Selective amplification is carried out using blocking property oligonucleotides
CN110892485A (en) 2017-02-22 2020-03-17 特韦斯特生物科学公司 Nucleic acid based data storage
US10894959B2 (en) 2017-03-15 2021-01-19 Twist Bioscience Corporation Variant libraries of the immunological synapse and synthesis thereof
AU2018281745B2 (en) 2017-06-05 2022-05-19 Becton, Dickinson And Company Sample indexing for single cells
WO2018226685A2 (en) 2017-06-06 2018-12-13 Dana-Farber Cancer Institute, Inc. Methods for sensitizing cancer cells to t cell-mediated killing by modulating molecular pathways
CA3066744A1 (en) 2017-06-12 2018-12-20 Twist Bioscience Corporation Methods for seamless nucleic acid assembly
WO2018231864A1 (en) 2017-06-12 2018-12-20 Twist Bioscience Corporation Methods for seamless nucleic acid assembly
WO2018231589A1 (en) 2017-06-14 2018-12-20 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Method for determining lymphoma type
KR20200047706A (en) 2017-09-11 2020-05-07 트위스트 바이오사이언스 코포레이션 GPCR binding protein and method for synthesis thereof
CA3075062A1 (en) 2017-09-11 2019-03-14 Cyteir Therapeutics, Inc. Rad51 inhibitors
CN111565834B (en) 2017-10-20 2022-08-26 特韦斯特生物科学公司 Heated nanopores for polynucleotide synthesis
US20200339987A1 (en) 2017-11-09 2020-10-29 Alnylam Pharmaceuticals, Inc. Assays and methods for determining expression of the lect2 gene
US20190142722A1 (en) 2017-11-10 2019-05-16 The Trustees Of Columbia University In The City Of New York Methods and compositions for promoting or inducing hair growth
WO2019109016A1 (en) 2017-12-01 2019-06-06 Millennium Pharmaceuticals, Inc. Biomarkers and methods for treatment with nae inhibitors
SG11202006460SA (en) 2018-01-04 2020-08-28 Twist Bioscience Corp Dna-based digital information storage
CN112567246A (en) 2018-01-25 2021-03-26 比奥根Ma公司 Method of treating spinal muscular atrophy
CN112243461A (en) 2018-05-03 2021-01-19 贝克顿迪金森公司 Molecular barcoding at opposite transcript ends
ES2945191T3 (en) 2018-05-03 2023-06-29 Becton Dickinson Co High-throughput multi-omics sample analysis
SG11202011467RA (en) 2018-05-18 2020-12-30 Twist Bioscience Corp Polynucleotides, reagents, and methods for nucleic acid hybridization
EP3861134A1 (en) 2018-10-01 2021-08-11 Becton, Dickinson and Company Determining 5' transcript sequences
ES2939143T3 (en) 2018-10-05 2023-04-19 Eisai R&D Man Co Ltd Biomarkers for a Combination Therapy Comprising Lenvatinib and Everolimus
EP3861346A1 (en) 2018-10-05 2021-08-11 Eisai R&D Management Co., Ltd. Biomarkers for a therapy comprising a sorafenib compound
EP3894552A1 (en) 2018-12-13 2021-10-20 Becton, Dickinson and Company Selective extension in single cell whole transcriptome analysis
US11371076B2 (en) 2019-01-16 2022-06-28 Becton, Dickinson And Company Polymerase chain reaction normalization through primer titration
EP4242322A3 (en) 2019-01-23 2023-09-20 Becton, Dickinson and Company Oligonucleotides associated with antibodies
US20220128578A1 (en) 2019-02-12 2022-04-28 Biogen Ma Inc. Biomarkers of progressive multifocal leukoencephalopathy
SG11202109322TA (en) 2019-02-26 2021-09-29 Twist Bioscience Corp Variant nucleic acid libraries for glp1 receptor
WO2020176680A1 (en) 2019-02-26 2020-09-03 Twist Bioscience Corporation Variant nucleic acid libraries for antibody optimization
SG11202109399SA (en) 2019-03-12 2021-09-29 Cyteir Therapeutics Inc Rad51 inhibitors
TW202102215A (en) 2019-03-25 2021-01-16 美商賽堤爾醫療公司 Combinations of rad51 and parp inhibitors
US11332738B2 (en) 2019-06-21 2022-05-17 Twist Bioscience Corporation Barcode-based nucleic acid sequence assembly
JP2022537372A (en) 2019-06-21 2022-08-25 サイティアー セラピューティクス,インコーポレイティド Methods of using RAD51 inhibitors to treat pancreatic cancer
CA3154157A1 (en) 2019-09-13 2021-03-18 23Andme, Inc. Methods and systems for determining and displaying pedigrees
JP2023500679A (en) 2019-11-08 2023-01-10 ベクトン・ディキンソン・アンド・カンパニー Using random priming to obtain full-length V(D)J information for immune repertoire sequencing
EP4090763A1 (en) 2020-01-13 2022-11-23 Becton Dickinson and Company Methods and compositions for quantitation of proteins and rna
CN115551502A (en) 2020-03-03 2022-12-30 赛泰尔治疗公司 Specific doses of RAD51 inhibitor compound 67A (2301085-06-1) for the treatment of cancer
EP4143333A1 (en) 2020-04-30 2023-03-08 Dimensiongen Devices and methods for macromolecular manipulation
EP4150118A1 (en) 2020-05-14 2023-03-22 Becton Dickinson and Company Primers for immune repertoire profiling
WO2022035729A1 (en) 2020-08-10 2022-02-17 Dimensiongen Devices and methods for multi-dimensional genome analysis
US20240024377A1 (en) 2020-10-29 2024-01-25 Evelo Biosciences, Inc. Compositions comprising spirulina components
US11739443B2 (en) 2020-11-20 2023-08-29 Becton, Dickinson And Company Profiling of highly expressed and lowly expressed proteins
CN117425674A (en) 2021-03-02 2024-01-19 丹娜法伯癌症研究院 Methods of treating red blood cell disorders
EP4074820A1 (en) 2021-04-16 2022-10-19 The Trustees of The University of Pennsylvania Micro-engineered models of the human eye and methods of use
WO2022251166A2 (en) 2021-05-25 2022-12-01 Evelo Biosciences, Inc. Bacterial compositions comprising soy hemoglobin
WO2022261183A2 (en) 2021-06-08 2022-12-15 Dana-Farber Cancer Institute, Inc. Compositions and methods for treating and/or identifying an agent for treating intestinal cancers
WO2023097119A2 (en) 2021-11-29 2023-06-01 Dana-Farber Cancer Institute, Inc. Methods and compositions to modulate riok2
GB202208171D0 (en) 2022-06-01 2022-07-13 Institute Of Cancer Res Method

Citations (93)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3077780A (en) * 1961-11-21 1963-02-19 Metrimpex Magyar Mueszeripari Volumetric liquid-transfer device
US3642450A (en) * 1969-01-16 1972-02-15 Kabi Ab Test strip packaged-unit
US3646346A (en) * 1968-12-26 1972-02-29 Pharmacia Ab Antibody-coated tube system for radioimmunoassay
US3861886A (en) * 1968-11-13 1975-01-21 Melpar Inc Material identification coding methods and systems
US4001583A (en) * 1974-10-04 1977-01-04 Barrett M James Covalently bound biological substances to plastic materials and use in radioassay
US4016855A (en) * 1974-09-04 1977-04-12 Hitachi, Ltd. Grinding method
US4086254A (en) * 1977-04-13 1978-04-25 The Upjohn Company Photocleavable steroids
US4145406A (en) * 1977-04-11 1979-03-20 Miles Laboratories, Inc. Specific binding - adsorbent assay method and test means
US4204929A (en) * 1978-04-18 1980-05-27 University Patents, Inc. Isoelectric focusing method
US4258001A (en) * 1978-12-27 1981-03-24 Eastman Kodak Company Element, structure and method for the analysis or transport of liquids
US4259223A (en) * 1976-03-29 1981-03-31 California Institute Of Technology Cross-linked polyvinyl pyridine coated glass particle catalyst support and aqueous composition or polyvinyl pyridine adducted microspheres
US4263504A (en) * 1979-08-01 1981-04-21 Ncr Corporation High density matrix code
US4267234A (en) * 1978-03-17 1981-05-12 California Institute Of Technology Polyglutaraldehyde synthesis and protein bonding substrates
US4325913A (en) * 1980-10-20 1982-04-20 Coulter Electronics, Inc. Reagent probe and method for fabrication thereof
US4378333A (en) * 1980-12-11 1983-03-29 Laipply Thomas C Device for preparing blood smears on glass slides and method therefor
US4427415A (en) * 1979-01-05 1984-01-24 Cleveland Patrick H Manifold vacuum biochemical test method and device
US4430299A (en) * 1981-06-18 1984-02-07 Coulter Electronics, Inc. Apparatus for monitoring chemical reactions
US4563417A (en) * 1984-08-31 1986-01-07 Miles Laboratories, Inc. Nucleic acid hybridization assay employing antibodies to intercalation complexes
US4586546A (en) * 1984-10-23 1986-05-06 Cetus Corporation Liquid handling device and method
US4591570A (en) * 1983-02-02 1986-05-27 Centocor, Inc. Matrix of antibody-coated spots for determination of antigens
US4647544A (en) * 1984-06-25 1987-03-03 Nicoli David F Immunoassay using optical interference detection
US4652533A (en) * 1983-04-28 1987-03-24 Pandex Laboratories, Inc. Method of solid phase immunoassay incorporating a luminescent label
US4654165A (en) * 1985-04-16 1987-03-31 Micro Tracers, Inc. Microingredient containing tracer
US4656007A (en) * 1984-10-01 1987-04-07 Commissariat A L'energie Atomique Programmable automatic means for the deposition in a precise position of a minute, precise quantity of liquid on an analytical support
US4668476A (en) * 1984-03-23 1987-05-26 Applied Biosystems, Inc. Automated polypeptide synthesis apparatus
US4719087A (en) * 1985-07-01 1988-01-12 Baxter Travenol Laboratories, Inc. Tray for analyzing system
US4737454A (en) * 1983-07-14 1988-04-12 Molecular Diagnostics, Inc. Fast photochemical method of labelling nucleic acids for detection purposes in hybridization assays
US4738669A (en) * 1984-08-24 1988-04-19 Vlock D G Fluid dispenser using capillary action
US4746490A (en) * 1983-09-22 1988-05-24 Saneii Hossain H Solid phase peptide synthesizer
US4797355A (en) * 1985-06-13 1989-01-10 Amgen Inc. Methods for attaching polynucleotides to supports
US4798706A (en) * 1985-09-13 1989-01-17 Fisher Scientific Co. Device for holding horizontal array of liquid aliquots
US4802101A (en) * 1985-08-19 1989-01-31 Fuji Photo Film Co., Ltd. Signal processing method for determining base sequence of nucleic acid
US4806631A (en) * 1985-09-30 1989-02-21 Miles Inc. Immobilization of nucleic acids on solvolyzed nylon supports
US4806546A (en) * 1985-09-30 1989-02-21 Miles Inc. Immobilization of nucleic acids on derivatized nylon supports
US4811218A (en) * 1986-06-02 1989-03-07 Applied Biosystems, Inc. Real time scanning electrophoresis apparatus for DNA sequencing
US4818492A (en) * 1986-03-20 1989-04-04 Kabushiki Kaisha Toshiba Capacitive liquid level sensor for automatic chemical analyzer
US4820812A (en) * 1982-08-09 1989-04-11 Wakunaga Seiyaku Kabushiki Kaisha Oligonucleotide derivatives and production thereof
US4822681A (en) * 1986-01-29 1989-04-18 Veb Leipziger Arzeimittelwerk Activated polymer solid bodies and processes for the production thereof
US4828801A (en) * 1985-07-02 1989-05-09 Centre National De La Recherche Scientifique Device for detecting on a nitrocellulose sheet the presence of macromolecular complexes, such as antigens/antibodies
US4894796A (en) * 1986-03-17 1990-01-16 Westinghouse Electric Corp. Automatic transfer switch with programmable display
US4906439A (en) * 1986-03-25 1990-03-06 Pb Diagnostic Systems, Inc. Biological diagnostic device and method of use
US4996646A (en) * 1988-03-31 1991-02-26 Square D Company Microprocessor-controlled circuit breaker and system
US4996142A (en) * 1986-09-04 1991-02-26 Agricultural Genetics Company Limited Non-radioactive nucleic acid hybridization probes
US4997278A (en) * 1987-08-22 1991-03-05 Amersham International Plc Biological sensors
US4997521A (en) * 1987-05-20 1991-03-05 Massachusetts Institute Of Technology Electrostatic micromotor
US5094594A (en) * 1990-04-23 1992-03-10 Genomyx, Incorporated Piezoelectric pumping device
US5196305A (en) * 1989-09-12 1993-03-23 Eastman Kodak Company Diagnostic and amplification methods using primers having thymine at 3' end to overcome primer-target mismatch at the 3' end
US5204268A (en) * 1988-06-02 1993-04-20 Fuji Photo Film Co., Ltd. Method and apparatus for applying liquid samples
US5279558A (en) * 1989-06-16 1994-01-18 Science Incorporated Fluid delivery apparatus with an additive
US5288514A (en) * 1992-09-14 1994-02-22 The Regents Of The University Of California Solid phase and combinatorial synthesis of benzodiazepine compounds on a solid support
US5291763A (en) * 1991-09-18 1994-03-08 Cuisinot Charles P Universal lock for quick release mechanism
US5300779A (en) * 1985-08-05 1994-04-05 Biotrack, Inc. Capillary flow device
US5306510A (en) * 1988-01-14 1994-04-26 Cyberlab, Inc. Automated pipetting system
US5306618A (en) * 1986-07-02 1994-04-26 E. I. Du Pont De Nemours And Company Method systems and reagents for DNA sequencing
US5378638A (en) * 1990-08-02 1995-01-03 Boehringer Mannheim Gmbh Analysis element and process for its manufacture
US5380489A (en) * 1992-02-18 1995-01-10 Eastman Kodak Company Element and method for nucleic acid amplification and detection using adhered probes
US5382512A (en) * 1993-08-23 1995-01-17 Chiron Corporation Assay device with captured particle reagent
US5384262A (en) * 1990-11-30 1995-01-24 Quantix Systems, L.P. Quantitative immunoassay for volatile organic compounds
US5482867A (en) * 1989-11-13 1996-01-09 Affymax Technologies N.V. Spatially-addressable immobilization of anti-ligands on surfaces
US5484908A (en) * 1991-11-26 1996-01-16 Gilead Sciences, Inc. Oligonucleotides containing 5-propynyl pyrimidines
US5486452A (en) * 1981-04-29 1996-01-23 Ciba-Geigy Corporation Devices and kits for immunological analysis
US5491074A (en) * 1993-04-01 1996-02-13 Affymax Technologies Nv Association peptides
US5491570A (en) * 1991-07-26 1996-02-13 Accuwave Corporation Methods and devices for using photorefractive materials at infrared wavelengths
US5491224A (en) * 1990-09-20 1996-02-13 Bittner; Michael L. Direct label transaminated DNA probe compositions for chromosome identification and methods for their manufacture
US5492840A (en) * 1988-11-10 1996-02-20 Pharmacia Biosensor Ab Surface plasmon resonance sensor unit and its use in biosensor systems
US5498530A (en) * 1991-10-16 1996-03-12 Affymax Technologies, N.V. Peptide library and screening method
US5503805A (en) * 1993-11-02 1996-04-02 Affymax Technologies N.V. Apparatus and method for parallel coupling reactions
US5599720A (en) * 1982-08-27 1997-02-04 Multilyte Limited Measurement of analyte concentration
US5605793A (en) * 1994-02-17 1997-02-25 Affymax Technologies N.V. Methods for in vitro recombination
US5607691A (en) * 1992-06-12 1997-03-04 Affymax Technologies N.V. Compositions and methods for enhanced drug delivery
US5608035A (en) * 1994-02-02 1997-03-04 Affymax Technologies N.V. Peptides and compounds that bind to the IL-1 receptor
US5723584A (en) * 1993-07-30 1998-03-03 Affymax Technologies N.V. Biotinylation of proteins
US5728802A (en) * 1992-05-06 1998-03-17 Affymax Technologies N.V. Peptides and compounds that bind selectins including endothelium leukocyte adhesion molecule 1 (ELAM-1)
US5733731A (en) * 1991-10-16 1998-03-31 Affymax Technologies N.V. Peptide library and screening method
US5861476A (en) * 1994-02-02 1999-01-19 Affymax Technologies N.V. Peptides and compounds that bind to the IL-1 receptor
US5871928A (en) * 1989-06-07 1999-02-16 Fodor; Stephen P. A. Methods for nucleic acid analysis
US5880096A (en) * 1994-02-02 1999-03-09 Affymax Technologies N.V. Peptides and compounds that bind to the IL-1 receptor
US5885837A (en) * 1991-11-22 1999-03-23 Affymetrix, Inc. Very large scale immobilized polymer synthesis using mechanically directed flow paths
US5889165A (en) * 1989-06-07 1999-03-30 Affymetrix, Inc. Photolabile nucleoside protecting groups
US5896047A (en) * 1997-02-05 1999-04-20 Xilinx, Inc. Balanced truth-and-complement circuit
US6040193A (en) * 1991-11-22 2000-03-21 Affymetrix, Inc. Combinatorial strategies for polymer synthesis
US6171797B1 (en) * 1999-10-20 2001-01-09 Agilent Technologies Inc. Methods of making polymeric arrays
US6197506B1 (en) * 1989-06-07 2001-03-06 Affymetrix, Inc. Method of detecting nucleic acids
US6368874B1 (en) * 1995-12-22 2002-04-09 Affymax, Inc. Methods for hard-tagging an encoded synthetic library
US6379895B1 (en) * 1989-06-07 2002-04-30 Affymetrix, Inc. Photolithographic and other means for manufacturing arrays
US20030003475A1 (en) * 1989-06-07 2003-01-02 Affymetrix, Inc. Arrays for detecting nucleic acids
US6506558B1 (en) * 1990-03-07 2003-01-14 Affymetrix Inc. Very large scale immobilized polymer synthesis
US20040029115A9 (en) * 1990-12-06 2004-02-12 Affymax Technologies, N.V. Sequencing of surface immobilized polymers utilizing microfluorescence detection
US6849462B1 (en) * 1991-11-22 2005-02-01 Affymetrix, Inc. Combinatorial strategies for polymer synthesis
US6852488B2 (en) * 1993-06-25 2005-02-08 Affymetrix, Inc. Identifying a base in a nucleic acid
US6864048B2 (en) * 1993-04-28 2005-03-08 Affymetrix, Inc. Factorial chemical libraries
US20050079529A1 (en) * 1989-06-07 2005-04-14 Affymetrix, Inc. Very large scale immobilized polymer synthesis
US7015046B2 (en) * 1995-03-10 2006-03-21 Mesoscale Technologies, Llc. Multi-array, multi-specific electrochemiluminescence testing

Family Cites Families (263)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US559720A (en) * 1896-05-05 Island
US3862056A (en) 1967-12-15 1975-01-21 Allied Chem Semiconductor polymer compositions comprising a grafted block copolymer of synthetic rubber and polyolefin and carbon black
US3730884A (en) * 1971-04-02 1973-05-01 B Burns Method and apparatus for conveying sewage
US3730844A (en) * 1971-08-27 1973-05-01 Purdue Research Foundation Polynucleotide analysis
DE2150691C2 (en) 1971-10-12 1982-09-09 Basf Ag, 6700 Ludwigshafen Photosensitive mixture and use of a photosensitive mixture for the production of a planographic printing plate
DE2242394A1 (en) 1972-08-29 1974-03-14 Basf Ag MIXTURES OF SUBSTANCE HARDLED UNDER THE EFFECT OF LIGHT
US3939350A (en) 1974-04-29 1976-02-17 Board Of Trustees Of The Leland Stanford Junior University Fluorescent immunoassay employing total reflection for activation
SE407115B (en) 1975-10-06 1979-03-12 Kabi Ab PROCEDURES AND METAL ELECTRODES FOR THE STUDY OF ENZYMATIC AND OTHER BIOCHEMICAL REACTIONS
US4238757A (en) 1976-03-19 1980-12-09 General Electric Company Field effect transistor for detection of biological reactions
US4121222A (en) * 1977-09-06 1978-10-17 A. B. Dick Company Drop counter ink replenishing system
US4180739A (en) 1977-12-23 1979-12-25 Varian Associates, Inc. Thermostatable flow cell for fluorescence measurements
US4448534A (en) 1978-03-30 1984-05-15 American Hospital Corporation Antibiotic susceptibility testing
US4269933A (en) 1978-06-08 1981-05-26 E. I. Du Pont De Nemours And Company Methods of developing photopolymerizable compositions containing an 0-nitroaromatic compound as photoinhibitor
US4216245A (en) * 1978-07-25 1980-08-05 Miles Laboratories, Inc. Method of making printed reagent test devices
US4314821A (en) 1979-04-09 1982-02-09 Minnesota Mining And Manufacturing Company Sandwich immunoassay using piezoelectric oscillator
US4342905A (en) 1979-08-31 1982-08-03 Nippon Kogaku K.K. Automatic focusing device of a microscope
US4500707A (en) 1980-02-29 1985-02-19 University Patents, Inc. Nucleosides useful in the preparation of polynucleotides
US4458066A (en) 1980-02-29 1984-07-03 University Patents, Inc. Process for preparing polynucleotides
US4327073A (en) 1980-04-07 1982-04-27 Huang Henry V Automated method for quantitative analysis of biological fluids
US4444892A (en) 1980-10-20 1984-04-24 Malmros Mark K Analytical device having semiconductive organic polymeric element associated with analyte-binding substance
IL61351A (en) 1980-10-27 1983-12-30 Yeda Res & Dev Organometallic polymers,their preparation and compositions of co-ordination compounds containing them
US4711955A (en) 1981-04-17 1987-12-08 Yale University Modified nucleotides and methods of preparing and using same
US4373071A (en) 1981-04-30 1983-02-08 City Of Hope Research Institute Solid-phase synthesis of polynucleotides
US4339528A (en) 1981-05-19 1982-07-13 Rca Corporation Etching method using a hardened PVA stencil
JPS57192856A (en) * 1981-05-25 1982-11-27 Ngk Insulators Ltd Oxygen concentration detector
US4732847A (en) * 1981-06-09 1988-03-22 University Of Hawaii Monoclonal antibodies for DNA-RNA hybrid complexes and their uses
US4498774A (en) * 1981-07-22 1985-02-12 Iowa State University Research Foundation, Inc. Micro-polarimeter for high performance liquid chromatography
US5087558A (en) * 1981-09-25 1992-02-11 Webster John A Jr Method for identifying and characterizing organisms
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
US4444878A (en) 1981-12-21 1984-04-24 Boston Biomedical Research Institute, Inc. Bispecific antibody determinants
JPS58189558A (en) 1982-04-28 1983-11-05 Mochida Pharmaceut Co Ltd Vessel for immunological measurement
US4500919A (en) * 1982-05-04 1985-02-19 Massachusetts Institute Of Technology Color reproduction system
US4483920A (en) 1982-05-17 1984-11-20 Hahnemann University Immobilization of message RNA directly from cells onto filter material
US4556643A (en) * 1982-07-26 1985-12-03 Agracetus Assay method and probe for polynucleotide sequences
US4624915A (en) 1982-07-29 1986-11-25 Board Of Trustees Of Michigan State University Positive selection sorting of cells
US4973493A (en) 1982-09-29 1990-11-27 Bio-Metric Systems, Inc. Method of improving the biocompatibility of solid surfaces
US4722906A (en) 1982-09-29 1988-02-02 Bio-Metric Systems, Inc. Binding reagents and methods
US4845027B1 (en) * 1982-10-13 1994-05-10 Biowhittaker Inc Fluorometric assay of allergic reactions
US4588682A (en) * 1982-12-13 1986-05-13 Integrated Genetics, Inc. Binding nucleic acid to a support
CA1237645A (en) * 1982-12-21 1988-06-07 John H. Fisher Assay technique
US4516833A (en) 1982-12-27 1985-05-14 University Of Dayton Production of high performance optical spatial filters
US4487839A (en) * 1983-01-05 1984-12-11 Ortho Diagnostic Systems Inc. Immunoassay methods employing patterns for the detection of soluble and cell surface antigens
DE3301833A1 (en) 1983-01-20 1984-07-26 Gesellschaft für Biotechnologische Forschung mbH (GBF), 3300 Braunschweig METHOD FOR SIMULTANEOUS SYNTHESIS OF SEVERAL OLIGONOCLEOTIDES IN A SOLID PHASE
US4994373A (en) 1983-01-27 1991-02-19 Enzo Biochem, Inc. Method and structures employing chemically-labelled polynucleotide probes
US4575433A (en) * 1983-01-31 1986-03-11 Iowa State University Research Foundation, Inc. Method of generating chemiluminescent light
US4634574A (en) * 1983-01-31 1987-01-06 Iowa State Research Foundation, Inc. Apparatus for use in sulfide chemiluminescence detection
US4537861A (en) 1983-02-03 1985-08-27 Elings Virgil B Apparatus and method for homogeneous immunoassay
US4584277A (en) 1983-04-05 1986-04-22 Syntex (U.S.A.) Inc. Fluorescent multiparameter particle analysis
GB8311018D0 (en) * 1983-04-22 1983-05-25 Amersham Int Plc Detecting mutations in dna
US4533682A (en) * 1983-04-29 1985-08-06 Desoto, Inc. Imidazolidine-blocked amine polymers
GB8314523D0 (en) 1983-05-25 1983-06-29 Lowe C R Diagnostic device
US4517338A (en) 1983-06-20 1985-05-14 Chiron Corporation Multiple reactor system and method for polynucleotide synthesis
US4542102A (en) 1983-07-05 1985-09-17 Molecular Diagnostics, Inc. Coupling of nucleic acids to solid support by photochemical methods
US4987065A (en) 1983-07-05 1991-01-22 Enzo Biochem, Inc. In vivo labelling of polynucleotide sequences
US4713326A (en) 1983-07-05 1987-12-15 Molecular Diagnostics, Inc. Coupling of nucleic acids to solid support by photochemical methods
US4626684A (en) 1983-07-13 1986-12-02 Landa Isaac J Rapid and automatic fluorescence immunoassay analyzer for multiple micro-samples
US5200313A (en) * 1983-08-05 1993-04-06 Miles Inc. Nucleic acid hybridization assay employing detectable anti-hybrid antibodies
US4677054A (en) 1983-08-08 1987-06-30 Sloan-Kettering Institute For Cancer Research Method for simple analysis of relative nucleic acid levels in multiple small samples by cytoplasmic dot hybridization
US4719615A (en) 1983-08-22 1988-01-12 Optical Data, Inc. Erasable optical data storage medium
US4598049A (en) * 1983-08-31 1986-07-01 Systec Inc. General purpose gene synthesizer
US4569967A (en) 1983-10-24 1986-02-11 The Salk Institute For Biological Studies Synthesis of N-substituted peptide amides
US4580895A (en) 1983-10-28 1986-04-08 Dynatech Laboratories, Incorporated Sample-scanning photometer
US4849513A (en) 1983-12-20 1989-07-18 California Institute Of Technology Deoxyribonucleoside phosphoramidites in which an aliphatic amino group is attached to the sugar ring and their use for the preparation of oligonucleotides containing aliphatic amino groups
US5821058A (en) * 1984-01-16 1998-10-13 California Institute Of Technology Automated DNA sequencing technique
US4613566A (en) 1984-01-23 1986-09-23 President And Fellows Of Harvard College Hybridization assay and kit therefor
JPS60155942A (en) * 1984-01-25 1985-08-16 Fuji Photo Film Co Ltd Spot deposition apparatus of liquid specimen
FI71768C (en) 1984-02-17 1987-02-09 Orion Yhtymae Oy Enhanced nucleic acid reagents and process for their preparation.
GB8405437D0 (en) * 1984-03-01 1984-04-04 Amersham Int Plc Detecting polynucleotide sequences
US4704353A (en) 1984-04-27 1987-11-03 Molecular Devices Corporation Photoresponsive redox detection and discrimination
EP0164206B1 (en) 1984-05-02 1988-11-02 Brendan James Hamill An apparatus for the chemical synthesis of oligonucleotides
US4766062A (en) * 1984-05-07 1988-08-23 Allied Corporation Displacement polynucleotide assay method and polynucleotide complex reagent therefor
US4670380A (en) * 1984-05-23 1987-06-02 Molecular Diagnostics, Inc. Assays utilizing labeled nucleic acid probes
US4555490A (en) 1984-06-08 1985-11-26 The United States Of America As Represented By The Department Of Health And Human Services Rapid visualization system for gel electrophoresis
US4755458A (en) 1984-08-30 1988-07-05 Enzo Biochem, Inc. Composition and method for the detection of the presence of a polynucleotide sequence of interest
US4681859A (en) 1984-09-21 1987-07-21 Ortho Diagnostic Systems Inc. Fluorescence polarization immunoassay for heavy antigens
US5258506A (en) 1984-10-16 1993-11-02 Chiron Corporation Photolabile reagents for incorporation into oligonucleotide chains
DE3440141A1 (en) 1984-11-02 1986-05-07 Heiner Dipl.-Chem. Dr. 8000 München Eckert Use of bistrichloromethyl carbonate as a proreagent for phosgene
US4820630A (en) 1984-11-23 1989-04-11 Digene Diagnostics, Incorporated Assay for nucleic acid sequences, particularly genetic lesions, using interactive labels
US4719179A (en) * 1984-11-30 1988-01-12 Pharmacia P-L Biochemicals, Inc. Six base oligonucleotide linkers and methods for their use
US5064754A (en) * 1984-12-14 1991-11-12 Mills Randell L Genomic sequencing method
US5026840A (en) 1985-01-10 1991-06-25 Molecular Diagnostics, Inc. Photochemical nucleic acid-labeling reagent having a polyalklamine spacer
US4713347A (en) 1985-01-14 1987-12-15 Sensor Diagnostics, Inc. Measurement of ligand/anti-ligand interactions using bulk conductance
US4767700A (en) * 1985-02-15 1988-08-30 Beckman Research Institute Of The City Of Hope Detection of particular nucleotide sequences
US5096807A (en) * 1985-03-06 1992-03-17 Murex Corporation Imaging immunoassay detection system with background compensation and its use
US5217866A (en) 1985-03-15 1993-06-08 Anti-Gene Development Group Polynucleotide assay reagent and method
US4631211A (en) 1985-03-25 1986-12-23 Scripps Clinic & Research Foundation Means for sequential solid phase organic synthesis and methods using the same
US4683202A (en) 1985-03-28 1987-07-28 Cetus Corporation Process for amplifying nucleic acid sequences
US6040166A (en) * 1985-03-28 2000-03-21 Roche Molecular Systems, Inc. Kits for amplifying and detecting nucleic acid sequences, including a probe
US4965188A (en) 1986-08-22 1990-10-23 Cetus Corporation Process for amplifying, detecting, and/or cloning nucleic acid sequences using a thermostable enzyme
US4683195A (en) * 1986-01-30 1987-07-28 Cetus Corporation Process for amplifying, detecting, and/or-cloning nucleic acid sequences
DE3513168A1 (en) 1985-04-12 1986-10-16 Thomas 8000 München Dandekar BIOSENSOR CONSISTING OF A SEMICONDUCTOR BASED ON SILICON OR CARBON-BASED (ELECTRONIC PART) AND NUCLEIN BASE (OR. OTHER BIOL. MONOMERS)
EP0199327B1 (en) * 1985-04-19 1992-03-04 Fuji Photo Film Co., Ltd. Signal processing method for determining base sequence of nucleic acids
NZ215865A (en) 1985-04-22 1988-10-28 Commw Serum Lab Commission Method of determining the active site of a receptor-binding analogue
FR2583772B1 (en) 1985-06-20 1987-08-28 Roussel Uclaf NEW MEDIA, THE PREPARATION OF SUCH MEDIA, THE NEW INTERMEDIATES OBTAINED, THEIR APPLICATION TO THE SYNTHESIS OF OLIGONUCLEOTIDES AND THE NEW NUCLEOSIDES AND OLIGONUCLEOTIDES RELATED TO THE MEDIA THUS OBTAINED
FR2584090B1 (en) 1985-06-27 1987-08-28 Roussel Uclaf NEW SUPPORTS, THEIR PREPARATION AND THE INTERMEDIATES OBTAINED, THEIR APPLICATION TO THE SYNTHESIS OF OLIGONUCLEOTIDES AND THE NEW NUCLEOSIDES AND OLIGONUCLEOTIDES RELATED TO THE SUPPORTS OBTAINED
EP0212807A3 (en) 1985-07-15 1988-01-07 Imperial Chemical Industries Plc Coloured compound containing a cationic group and a reactive group
DD254998A1 (en) 1985-07-26 1988-03-16 Zeiss Jena Veb Carl ARRANGEMENT FOR THE IMAGE AND ANALYSIS OF FLUORESCENCE SIGNALS
WO1987000926A1 (en) 1985-08-05 1987-02-12 Rijksuniversiteit Leiden Labelled macromolecules; a process for their preparation and their use for immunological or immunocytochemical assays
EP0213825A3 (en) * 1985-08-22 1989-04-26 Molecular Devices Corporation Multiple chemically modulated capacitance
US4806312A (en) * 1985-08-28 1989-02-21 Miles Inc. Multizone analytical element having detectable signal concentrating zone
US4725536A (en) * 1985-09-19 1988-02-16 Genetics Institute, Inc. Reagent polynucleotide complex with multiple target binding regions, and kit and methods
US4725537A (en) * 1985-09-19 1988-02-16 Allied Corporation Assay, reagent and kit employing nucleic acid strand displacement and restriction endonuclease cleavage
TW203120B (en) * 1985-10-04 1993-04-01 Abbott Lab
IT1206532B (en) * 1985-10-15 1989-04-27 Marcucci Francesco MUCOSAL ALLERGY-DIAGNOSTIC METHOD AND ITS DEVICE FOR IN VIVO SURVEYING OF SPECIFIC AND TOTAL IGE
SE450171B (en) * 1985-10-18 1987-06-09 Carl Urban Ungerstedt DEVICE FOR AUTOMATIC COLLECTION OF SMALL LIQUID VOLUMES
US4888278A (en) 1985-10-22 1989-12-19 University Of Massachusetts Medical Center In-situ hybridization to detect nucleic acid sequences in morphologically intact cells
US4741043B1 (en) * 1985-11-04 1994-08-09 Cell Analysis Systems Inc Method of and apparatus for image analyses of biological specimens
US4822566A (en) 1985-11-19 1989-04-18 The Johns Hopkins University Optimized capacitive sensor for chemical analysis and measurement
US4882269A (en) * 1985-12-13 1989-11-21 Princeton University Amplified hybridization assay
US4637861A (en) 1985-12-16 1987-01-20 Allied Corporation Stabilized, lipid membrane-based device and method of analysis
US5447841A (en) 1986-01-16 1995-09-05 The Regents Of The Univ. Of California Methods for chromosome-specific staining
US4855225A (en) 1986-02-07 1989-08-08 Applied Biosystems, Inc. Method of detecting electrophoretically separated oligonucleotides
JPH0685387B2 (en) 1986-02-14 1994-10-26 株式会社東芝 Alignment method
US4916056A (en) * 1986-02-18 1990-04-10 Abbott Laboratories Solid-phase analytical device and method for using same
US4868103A (en) 1986-02-19 1989-09-19 Enzo Biochem, Inc. Analyte detection by means of energy transfer
US5348855A (en) 1986-03-05 1994-09-20 Miles Inc. Assay for nucleic acid sequences in an unpurified sample
EP0240729A3 (en) * 1986-03-05 1988-08-24 Fuji Photo Film Co., Ltd. Method of analyzing autoradiograph for determining base sequence of nucleic acid
US4728591A (en) 1986-03-07 1988-03-01 Trustees Of Boston University Self-assembled nanometer lithographic masks and templates and method for parallel fabrication of nanometer scale multi-device structures
US4925785A (en) 1986-03-07 1990-05-15 Biotechnica Diagnostics, Inc. Nucleic acid hybridization assays
US5604099A (en) * 1986-03-13 1997-02-18 Hoffmann-La Roche Inc. Process for detecting specific nucleotide variations and genetic polymorphisms present in nucleic acids
US5567809A (en) 1986-03-13 1996-10-22 Hoffmann-La Roche Inc. Methods and reagents for HLA DRbeta DNA typing
US5310893A (en) 1986-03-31 1994-05-10 Hoffmann-La Roche Inc. Method for HLA DP typing
US5256549A (en) 1986-03-28 1993-10-26 Chiron Corporation Purification of synthetic oligomers
US5153319A (en) 1986-03-31 1992-10-06 University Patents, Inc. Process for preparing polynucleotides
US4846552A (en) 1986-04-16 1989-07-11 The United States Of America As Represented By The Secretary Of The Air Force Method of fabricating high efficiency binary planar optical elements
US4981783A (en) 1986-04-16 1991-01-01 Montefiore Medical Center Method for detecting pathological conditions
US4808508A (en) 1986-07-01 1989-02-28 Hoechst Celanese Corporation Negative working color proofing process comprising diazo compound and polyvinyl acetal/polyvinyl alcohol/polyvinyl acetate resin
US4786684A (en) 1986-08-21 1988-11-22 The Mount Sinai School Of Medicine Of The City University Of New York Benzylthioether-linked solid support-bound thiol compounds and method for peptide synthesis
US5021550A (en) 1986-10-07 1991-06-04 Thomas Jefferson University Method for preventing deletion sequences in solid phase synthesis
GB8624637D0 (en) 1986-10-14 1986-11-19 Emi Plc Thorn Electrical device
US4979959A (en) 1986-10-17 1990-12-25 Bio-Metric Systems, Inc. Biocompatible coating for solid surfaces
US4877745A (en) * 1986-11-17 1989-10-31 Abbott Laboratories Apparatus and process for reagent fluid dispensing and printing
US5028525A (en) 1986-11-24 1991-07-02 Regents Of The University Of California Method of preparing and applying single stranded DNA probes to double stranded target DNAs in situ
JPS63167313A (en) 1986-12-27 1988-07-11 Hitachi Ltd Automatic focus control method
JPH0664057B2 (en) * 1987-01-06 1994-08-22 富士写真フイルム株式会社 Signal processing method for autoradiographic analysis
US4762881A (en) 1987-01-09 1988-08-09 E. I. Du Pont De Nemours And Company Photoreactive benzoylphenylalanines and related peptides
US5077085A (en) * 1987-03-06 1991-12-31 Schnur Joel M High resolution metal patterning of ultra-thin films on solid substrates
US5079600A (en) 1987-03-06 1992-01-07 Schnur Joel M High resolution patterning on solid substrates
US5037882A (en) * 1987-03-11 1991-08-06 Steel Samuel L Synthesis of oligonucleotide analogs
US4794150A (en) 1987-03-11 1988-12-27 Samuel Steel Synthesis of peptide analogs
US5026773A (en) 1987-03-11 1991-06-25 Samuel Steel Apparatus for a solid phase synthesis of peptide analogs
US4829010A (en) * 1987-03-13 1989-05-09 Tanox Biosystems, Inc. Immunoassay device enclosing matrixes of antibody spots for cell determinations
US5525464A (en) 1987-04-01 1996-06-11 Hyseq, Inc. Method of sequencing by hybridization of oligonucleotide probes
US5202231A (en) 1987-04-01 1993-04-13 Drmanac Radoje T Method of sequencing of genomes by hybridization of oligonucleotide probes
US6270961B1 (en) * 1987-04-01 2001-08-07 Hyseq, Inc. Methods and apparatus for DNA sequencing and DNA identification
US5100777A (en) * 1987-04-27 1992-03-31 Tanox Biosystems, Inc. Antibody matrix device and method for evaluating immune status
SE458968B (en) * 1987-06-16 1989-05-22 Wallac Oy BIOSPECIFIC ANALYTICAL PROCEDURE FOR MULTIPLE ANALYTICS WHICH DO NOT INCLUDE PARTICULAR COATING AND LABELING WITH FLUORESCING LABEL SUBSTANCES
US4880750A (en) * 1987-07-09 1989-11-14 Miragen, Inc. Individual-specific antibody identification methods
DE3722958A1 (en) * 1987-07-11 1989-01-19 Heinrich Dr Klefenz Method for the identification and/or isolation of specific molecules or populations of molecules
US4874500A (en) 1987-07-15 1989-10-17 Sri International Microelectrochemical sensor and sensor array
US4921805A (en) * 1987-07-29 1990-05-01 Life Technologies, Inc. Nucleic acid capture method
GB8803000D0 (en) * 1988-02-10 1988-03-09 Ekins Roger Philip Determination of ambient concentrations of several analytes
US5149625A (en) * 1987-08-11 1992-09-22 President And Fellows Of Harvard College Multiplex analysis of DNA
US4923901A (en) 1987-09-04 1990-05-08 Millipore Corporation Membranes with bound oligonucleotides and peptides
US5011770A (en) * 1987-09-04 1991-04-30 Molecular Devices, Inc. DNA detection method
US5089000A (en) * 1987-09-18 1992-02-18 John M. Agee Surgical method and instrument therefor
US5006464A (en) * 1987-10-01 1991-04-09 E-Y Laboratories, Inc. Directed flow diagnostic device and method
US4981985A (en) 1987-10-20 1991-01-01 Trustees Of The University Of Pennsylvania Synthesis of photolabile chelators for multivalent cations
US4886741A (en) 1987-12-09 1989-12-12 Microprobe Corporation Use of volume exclusion agents for the enhancement of in situ hybridization
US4844617A (en) 1988-01-20 1989-07-04 Tencor Instruments Confocal measuring microscope with automatic focusing
US5093245A (en) * 1988-01-26 1992-03-03 Applied Biosystems Labeling by simultaneous ligation and restriction
US5082830A (en) 1988-02-26 1992-01-21 Enzo Biochem, Inc. End labeled nucleotide probe
GB8810400D0 (en) * 1988-05-03 1988-06-08 Southern E Analysing polynucleotide sequences
US4946942A (en) 1988-03-11 1990-08-07 Bioresearch, Inc. Urethane-protected amino acid-N-carboxyanhydrides
US4986942A (en) * 1988-03-22 1991-01-22 Outboard Marine Corporation Method for forming ported cylinder sleeve liner foam pattern
US4988617A (en) 1988-03-25 1991-01-29 California Institute Of Technology Method of detecting a nucleotide change in nucleic acids
IL89964A0 (en) * 1988-04-15 1989-12-15 Montefiore Med Center Method for determining malignant progression,cdna,polypeptides encoded thereby and pharmaceutical compositions containing the same
US5002867A (en) 1988-04-25 1991-03-26 Macevicz Stephen C Nucleic acid sequence determination by multiple mixed oligonucleotide probes
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
US5013669A (en) * 1988-06-01 1991-05-07 Smithkline Diagnostics, Inc. Mass producible biologically active solid phase devices
US5047321A (en) * 1988-06-15 1991-09-10 Becton Dickinson & Co. Method for analysis of cellular components of a fluid
DE3923165A1 (en) 1988-07-15 1990-01-18 Hitachi Ltd MAGNETIC DISK DEVICE
US5094939A (en) * 1988-07-19 1992-03-10 Fujirebio, Inc. Chemiluminescence assays using stabilized dioxetane derivatives
US5075077A (en) 1988-08-02 1991-12-24 Abbott Laboratories Test card for performing assays
US5188963A (en) * 1989-11-17 1993-02-23 Gene Tec Corporation Device for processing biological specimens for analysis of nucleic acids
US5281540A (en) * 1988-08-02 1994-01-25 Abbott Laboratories Test array for performing assays
US4992383A (en) 1988-08-05 1991-02-12 Porton Instruments, Inc. Method for protein and peptide sequencing using derivatized glass supports
US5206137A (en) 1988-09-08 1993-04-27 Lifecodes Corporation Compositions and methods useful for genetic analysis
GB8822228D0 (en) * 1988-09-21 1988-10-26 Southern E M Support-bound oligonucleotides
FR2638848B1 (en) * 1988-11-04 1993-01-22 Chemunex Sa METHOD OF DETECTION AND / OR DETERMINATION IN A LIQUID OR SEMI-LIQUID MEDIUM OF AT LEAST ONE ORGANIC, BIOLOGICAL OR MEDICINAL SUBSTANCE, BY AN AGGLUTINATION METHOD
US5200051A (en) 1988-11-14 1993-04-06 I-Stat Corporation Wholly microfabricated biosensors and process for the manufacture and use thereof
US5869237A (en) * 1988-11-15 1999-02-09 Yale University Amplification karyotyping
US5215889A (en) 1988-11-18 1993-06-01 The Regents Of The University Of California Catalytic and reactive polypeptides and methods for their preparation and use
US5489507A (en) 1988-11-30 1996-02-06 Perkin-Elmer Corporation DNA detection by color complementation
US5047524A (en) 1988-12-21 1991-09-10 Applied Biosystems, Inc. Automated system for polynucleotide synthesis and purification
US5077210A (en) 1989-01-13 1991-12-31 Eigler Frances S Immobilization of active agents on substrates with a silane and heterobifunctional crosslinking agent
US4984857A (en) * 1989-01-17 1991-01-15 Iowa State University Research Foundation, Inc. Linearization of scan velocity of resonant vibrating-mirror beam deflectors
ATE123780T1 (en) 1989-02-17 1995-06-15 Chiron Mimotopes Pty Ltd METHOD FOR USING AND PRODUCING PEPTIDES.
US5081584A (en) 1989-03-13 1992-01-14 United States Of America Computer-assisted design of anti-peptides based on the amino acid sequence of a target peptide
EP0392546A3 (en) * 1989-04-14 1991-09-11 Ro Institut Za Molekularnu Genetiku I Geneticko Inzenjerstvo Process for determination of a complete or a partial contents of very short sequences in the samples of nucleic acids connected to the discrete particles of microscopic size by hybridization with oligonucleotide probes
US5112962A (en) 1989-04-19 1992-05-12 Northwestern University Labile anchors for solid phase polynucleotide synthesis
WO1990014441A1 (en) 1989-05-22 1990-11-29 Cetus Corporation Methods for tagging and tracing materials with nucleic acids
US5219726A (en) * 1989-06-02 1993-06-15 The Salk Institute For Biological Studies Physical mapping of complex genomes
US6309822B1 (en) * 1989-06-07 2001-10-30 Affymetrix, Inc. Method for comparing copy number of nucleic acid sequences
US5242974A (en) 1991-11-22 1993-09-07 Affymax Technologies N.V. Polymer reversal on solid surfaces
US6919211B1 (en) * 1989-06-07 2005-07-19 Affymetrix, Inc. Polypeptide 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
US5527681A (en) 1989-06-07 1996-06-18 Affymax Technologies N.V. Immobilized molecular synthesis of systematically substituted compounds
US5424186A (en) 1989-06-07 1995-06-13 Affymax Technologies N.V. Very large scale immobilized polymer synthesis
GB2233654A (en) 1989-07-07 1991-01-16 Nat Res Dev Immobilised polynucleotides
FR2649518B1 (en) * 1989-07-07 1991-10-18 Bioprobe Systems Sa HIGH SECURITY ENCRYPTED MARKING METHOD AND DEVICE FOR THE PROTECTION OF VALUABLE OBJECTS
DE3924454A1 (en) 1989-07-24 1991-02-07 Cornelis P Prof Dr Hollenberg THE APPLICATION OF DNA AND DNA TECHNOLOGY FOR THE CONSTRUCTION OF NETWORKS FOR USE IN CHIP CONSTRUCTION AND CHIP PRODUCTION (DNA CHIPS)
US5141813A (en) 1989-08-28 1992-08-25 Clontech Laboratories, Inc. Multifunctional controlled pore glass reagent for solid phase oligonucleotide synthesis
US5232829A (en) 1989-09-29 1993-08-03 Hoffmann-La Roche Inc. Detection of chlamydia trachomatis by polymerase chain reaction using biotin labelled lina primers and capture probes
US5297288A (en) * 1989-11-28 1994-03-22 United States Biochemical Corporation System for use with a high resolution scanner for scheduling a sequence of software tools for determining the presence of bands in DNA sequencing samples
US5215882A (en) 1989-11-30 1993-06-01 Ortho Diagnostic Systems, Inc. Method of immobilizing nucleic acid on a solid surface for use in nucleic acid hybridization assays
US5282490A (en) * 1989-12-18 1994-02-01 Higgs Robert E Flow metering injection controller
US5091652A (en) 1990-01-12 1992-02-25 The Regents Of The University Of California Laser excited confocal microscope fluorescence scanner and method
US5192407A (en) * 1990-01-30 1993-03-09 Iowa State University Research Foundation, Inc. Means and method of detection in chemical separation procedures
US5427908A (en) * 1990-05-01 1995-06-27 Affymax Technologies N.V. Recombinant library screening methods
US5494810A (en) * 1990-05-03 1996-02-27 Cornell Research Foundation, Inc. Thermostable ligase-mediated DNA amplifications system for the detection of genetic disease
ES2116977T3 (en) * 1990-05-11 1998-08-01 Microprobe Corp SOLID SUPPORTS FOR NUCLEIC ACID HYBRIDIZATION TESTS AND METHODS TO IMMOBILIZE OLIGONUCLEOTIDES IN A COVALENT WAY.
US5100626A (en) * 1990-05-24 1992-03-31 Levin Andrew E Binding assay device with removable cassette and manifold
US5723286A (en) * 1990-06-20 1998-03-03 Affymax Technologies N.V. Peptide library and screening systems
GB9014263D0 (en) 1990-06-27 1990-08-15 Dixon Arthur E Apparatus and method for spatially- and spectrally- resolvedmeasurements
US5306641A (en) 1990-07-27 1994-04-26 Saccocio Edward J Apparatus and method for determining gel rate of polymerizable compositions
US5235028A (en) 1990-08-31 1993-08-10 University Of Minnesota Polyethylene glycol derivatives for solid-phase applications
US6582908B2 (en) * 1990-12-06 2003-06-24 Affymetrix, Inc. Oligonucleotides
RU1794088C (en) 1991-03-18 1993-02-07 Институт Молекулярной Биологии Ан@ Ссср Method of dna nucleotide sequence determination and a device for its realization
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
DK0604552T3 (en) * 1991-09-18 1997-08-04 Affymax Tech Nv Process for the synthesis of different assemblies of oligomers
US5556961A (en) 1991-11-15 1996-09-17 Foote; Robert S. Nucleosides with 5'-O-photolabile protecting groups
US5846708A (en) 1991-11-19 1998-12-08 Massachusetts Institiute Of Technology Optical and electrical methods and apparatus for molecule detection
US6943034B1 (en) * 1991-11-22 2005-09-13 Affymetrix, Inc. Combinatorial strategies for polymer synthesis
US5412087A (en) 1992-04-24 1995-05-02 Affymax Technologies N.V. Spatially-addressable immobilization of oligonucleotides and other biological polymers on surfaces
US5324633A (en) 1991-11-22 1994-06-28 Affymax Technologies N.V. Method and apparatus for measuring binding affinity
US5573905A (en) 1992-03-30 1996-11-12 The Scripps Research Institute Encoded combinatorial chemical libraries
US5382514A (en) * 1992-03-31 1995-01-17 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services In vivo angiogenesis assay
US5667667A (en) 1992-04-24 1997-09-16 Isis Innovation Limited Electrochemical treatment of surfaces
GB9218131D0 (en) * 1992-08-26 1992-10-14 Slater James H A method of marking a liquid
US5565324A (en) 1992-10-01 1996-10-15 The Trustees Of Columbia University In The City Of New York Complex combinatorial chemical libraries encoded with tags
US5807683A (en) 1992-11-19 1998-09-15 Combichem, Inc. Combinatorial libraries and methods for their use
GB9315847D0 (en) 1993-07-30 1993-09-15 Isis Innovation Tag reagent and assay method
AU700315B2 (en) 1993-10-28 1998-12-24 Houston Advanced Research Center Microfabricated, flowthrough porous apparatus for discrete detection of binding reactions
US5591578A (en) * 1993-12-10 1997-01-07 California Institute Of Technology Nucleic acid mediated electron transfer
ZA95260B (en) 1994-01-13 1995-09-28 Univ Columbia Synthetic receptors libraries and uses thereof
US5571639A (en) 1994-05-24 1996-11-05 Affymax Technologies N.V. Computer-aided engineering system for design of sequence arrays and lithographic masks
US6287850B1 (en) * 1995-06-07 2001-09-11 Affymetrix, Inc. Bioarray chip reaction apparatus and its manufacture
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
US5727098A (en) * 1994-09-07 1998-03-10 Jacobson; Joseph M. Oscillating fiber optic display and imager
US5604097A (en) 1994-10-13 1997-02-18 Spectragen, Inc. Methods for sorting polynucleotides using oligonucleotide tags
US5846719A (en) 1994-10-13 1998-12-08 Lynx Therapeutics, Inc. Oligonucleotide tags for sorting and identification
US5695934A (en) 1994-10-13 1997-12-09 Lynx Therapeutics, Inc. Massively parallel sequencing of sorted polynucleotides
US5556752A (en) 1994-10-24 1996-09-17 Affymetrix, Inc. Surface-bound, unimolecular, double-stranded DNA
US6974666B1 (en) 1994-10-21 2005-12-13 Appymetric, Inc. Methods of enzymatic discrimination enhancement and surface-bound double-stranded DNA
US5830645A (en) 1994-12-09 1998-11-03 The Regents Of The University Of California Comparative fluorescence hybridization to nucleic acid arrays
US5776737A (en) * 1994-12-22 1998-07-07 Visible Genetics Inc. Method and composition for internal identification of samples
US5599695A (en) * 1995-02-27 1997-02-04 Affymetrix, Inc. Printing molecular library arrays using deprotection agents solely in the vapor phase
US5751629A (en) 1995-04-25 1998-05-12 Irori Remotely programmable matrices with memories
US5690894A (en) 1995-05-23 1997-11-25 The Regents Of The University Of California High density array fabrication and readout method for a fiber optic biosensor
US5707806A (en) 1995-06-07 1998-01-13 Genzyme Corporation Direct sequence identification of mutations by cleavage- and ligation-associated mutation-specific sequencing
US5777888A (en) 1995-08-09 1998-07-07 Regents Of The University Of California Systems for generating and analyzing stimulus-response output signal matrices
US5698393A (en) 1995-08-18 1997-12-16 Abbott Laboratories Method for elimination of rheumatoid factor interference in diagnostic assays
US5871697A (en) 1995-10-24 1999-02-16 Curagen Corporation Method and apparatus for identifying, classifying, or quantifying DNA sequences in a sample without sequencing
US5861071A (en) * 1995-11-21 1999-01-19 Alconex Specialty Products, Inc. Electrically insulated magnet wire and method of making the same
US5641634A (en) 1995-11-30 1997-06-24 Mandecki; Wlodek Electronically-indexed solid-phase assay for biomolecules
US6060240A (en) 1996-12-13 2000-05-09 Arcaris, Inc. Methods for measuring relative amounts of nucleic acids in a complex mixture and retrieval of specific sequences therefrom
US6023540A (en) 1997-03-14 2000-02-08 Trustees Of Tufts College Fiber optic sensor with encoded microspheres
US6306643B1 (en) * 1998-08-24 2001-10-23 Affymetrix, Inc. Methods of using an array of pooled probes in genetic analysis
US6346423B1 (en) * 1999-07-16 2002-02-12 Agilent Technologies, Inc. Methods and compositions for producing biopolymeric arrays
JP2004502705A (en) * 2000-07-03 2004-01-29 メルク エンド カムパニー インコーポレーテッド Coding methods in combinatorial libraries
US7492462B2 (en) * 2006-01-17 2009-02-17 Honeywell International, Inc. Optochemical sensor

Patent Citations (99)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3077780A (en) * 1961-11-21 1963-02-19 Metrimpex Magyar Mueszeripari Volumetric liquid-transfer device
US3861886A (en) * 1968-11-13 1975-01-21 Melpar Inc Material identification coding methods and systems
US3646346A (en) * 1968-12-26 1972-02-29 Pharmacia Ab Antibody-coated tube system for radioimmunoassay
US3642450A (en) * 1969-01-16 1972-02-15 Kabi Ab Test strip packaged-unit
US4016855A (en) * 1974-09-04 1977-04-12 Hitachi, Ltd. Grinding method
US4001583A (en) * 1974-10-04 1977-01-04 Barrett M James Covalently bound biological substances to plastic materials and use in radioassay
US4259223A (en) * 1976-03-29 1981-03-31 California Institute Of Technology Cross-linked polyvinyl pyridine coated glass particle catalyst support and aqueous composition or polyvinyl pyridine adducted microspheres
US4145406A (en) * 1977-04-11 1979-03-20 Miles Laboratories, Inc. Specific binding - adsorbent assay method and test means
US4086254A (en) * 1977-04-13 1978-04-25 The Upjohn Company Photocleavable steroids
US4267234A (en) * 1978-03-17 1981-05-12 California Institute Of Technology Polyglutaraldehyde synthesis and protein bonding substrates
US4204929A (en) * 1978-04-18 1980-05-27 University Patents, Inc. Isoelectric focusing method
US4258001A (en) * 1978-12-27 1981-03-24 Eastman Kodak Company Element, structure and method for the analysis or transport of liquids
US4427415A (en) * 1979-01-05 1984-01-24 Cleveland Patrick H Manifold vacuum biochemical test method and device
US4263504A (en) * 1979-08-01 1981-04-21 Ncr Corporation High density matrix code
US4325913A (en) * 1980-10-20 1982-04-20 Coulter Electronics, Inc. Reagent probe and method for fabrication thereof
US4378333A (en) * 1980-12-11 1983-03-29 Laipply Thomas C Device for preparing blood smears on glass slides and method therefor
US5486452A (en) * 1981-04-29 1996-01-23 Ciba-Geigy Corporation Devices and kits for immunological analysis
US4430299A (en) * 1981-06-18 1984-02-07 Coulter Electronics, Inc. Apparatus for monitoring chemical reactions
US4820812A (en) * 1982-08-09 1989-04-11 Wakunaga Seiyaku Kabushiki Kaisha Oligonucleotide derivatives and production thereof
US5599720A (en) * 1982-08-27 1997-02-04 Multilyte Limited Measurement of analyte concentration
US4591570A (en) * 1983-02-02 1986-05-27 Centocor, Inc. Matrix of antibody-coated spots for determination of antigens
US4652533A (en) * 1983-04-28 1987-03-24 Pandex Laboratories, Inc. Method of solid phase immunoassay incorporating a luminescent label
US4737454A (en) * 1983-07-14 1988-04-12 Molecular Diagnostics, Inc. Fast photochemical method of labelling nucleic acids for detection purposes in hybridization assays
US4746490A (en) * 1983-09-22 1988-05-24 Saneii Hossain H Solid phase peptide synthesizer
US4668476A (en) * 1984-03-23 1987-05-26 Applied Biosystems, Inc. Automated polypeptide synthesis apparatus
US4647544A (en) * 1984-06-25 1987-03-03 Nicoli David F Immunoassay using optical interference detection
US4738669A (en) * 1984-08-24 1988-04-19 Vlock D G Fluid dispenser using capillary action
US4563417A (en) * 1984-08-31 1986-01-07 Miles Laboratories, Inc. Nucleic acid hybridization assay employing antibodies to intercalation complexes
US4656007A (en) * 1984-10-01 1987-04-07 Commissariat A L'energie Atomique Programmable automatic means for the deposition in a precise position of a minute, precise quantity of liquid on an analytical support
US4586546A (en) * 1984-10-23 1986-05-06 Cetus Corporation Liquid handling device and method
US4654165A (en) * 1985-04-16 1987-03-31 Micro Tracers, Inc. Microingredient containing tracer
US4797355A (en) * 1985-06-13 1989-01-10 Amgen Inc. Methods for attaching polynucleotides to supports
US4719087A (en) * 1985-07-01 1988-01-12 Baxter Travenol Laboratories, Inc. Tray for analyzing system
US4828801A (en) * 1985-07-02 1989-05-09 Centre National De La Recherche Scientifique Device for detecting on a nitrocellulose sheet the presence of macromolecular complexes, such as antigens/antibodies
US5300779A (en) * 1985-08-05 1994-04-05 Biotrack, Inc. Capillary flow device
US4802101A (en) * 1985-08-19 1989-01-31 Fuji Photo Film Co., Ltd. Signal processing method for determining base sequence of nucleic acid
US4798706A (en) * 1985-09-13 1989-01-17 Fisher Scientific Co. Device for holding horizontal array of liquid aliquots
US4806546A (en) * 1985-09-30 1989-02-21 Miles Inc. Immobilization of nucleic acids on derivatized nylon supports
US4806631A (en) * 1985-09-30 1989-02-21 Miles Inc. Immobilization of nucleic acids on solvolyzed nylon supports
US4822681A (en) * 1986-01-29 1989-04-18 Veb Leipziger Arzeimittelwerk Activated polymer solid bodies and processes for the production thereof
US4894796A (en) * 1986-03-17 1990-01-16 Westinghouse Electric Corp. Automatic transfer switch with programmable display
US4818492A (en) * 1986-03-20 1989-04-04 Kabushiki Kaisha Toshiba Capacitive liquid level sensor for automatic chemical analyzer
US4906439A (en) * 1986-03-25 1990-03-06 Pb Diagnostic Systems, Inc. Biological diagnostic device and method of use
US4811218A (en) * 1986-06-02 1989-03-07 Applied Biosystems, Inc. Real time scanning electrophoresis apparatus for DNA sequencing
US5306618A (en) * 1986-07-02 1994-04-26 E. I. Du Pont De Nemours And Company Method systems and reagents for DNA sequencing
US4996142A (en) * 1986-09-04 1991-02-26 Agricultural Genetics Company Limited Non-radioactive nucleic acid hybridization probes
US4997521A (en) * 1987-05-20 1991-03-05 Massachusetts Institute Of Technology Electrostatic micromotor
US4997278A (en) * 1987-08-22 1991-03-05 Amersham International Plc Biological sensors
US5306510A (en) * 1988-01-14 1994-04-26 Cyberlab, Inc. Automated pipetting system
US4996646A (en) * 1988-03-31 1991-02-26 Square D Company Microprocessor-controlled circuit breaker and system
US5204268A (en) * 1988-06-02 1993-04-20 Fuji Photo Film Co., Ltd. Method and apparatus for applying liquid samples
US5492840A (en) * 1988-11-10 1996-02-20 Pharmacia Biosensor Ab Surface plasmon resonance sensor unit and its use in biosensor systems
US6355432B1 (en) * 1989-06-07 2002-03-12 Affymetrix Lnc. Products for detecting nucleic acids
US6379895B1 (en) * 1989-06-07 2002-04-30 Affymetrix, Inc. Photolithographic and other means for manufacturing arrays
US6197506B1 (en) * 1989-06-07 2001-03-06 Affymetrix, Inc. Method of detecting nucleic acids
US5889165A (en) * 1989-06-07 1999-03-30 Affymetrix, Inc. Photolabile nucleoside protecting groups
US5871928A (en) * 1989-06-07 1999-02-16 Fodor; Stephen P. A. Methods for nucleic acid analysis
US20030003475A1 (en) * 1989-06-07 2003-01-02 Affymetrix, Inc. Arrays for detecting nucleic acids
US20050079529A1 (en) * 1989-06-07 2005-04-14 Affymetrix, Inc. Very large scale immobilized polymer synthesis
US5279558A (en) * 1989-06-16 1994-01-18 Science Incorporated Fluid delivery apparatus with an additive
US5196305A (en) * 1989-09-12 1993-03-23 Eastman Kodak Company Diagnostic and amplification methods using primers having thymine at 3' end to overcome primer-target mismatch at the 3' end
US5482867A (en) * 1989-11-13 1996-01-09 Affymax Technologies N.V. Spatially-addressable immobilization of anti-ligands on surfaces
US20030017484A1 (en) * 1990-03-07 2003-01-23 Affymetrix, Inc. Arrays for detecting nucleic acids
US6506558B1 (en) * 1990-03-07 2003-01-14 Affymetrix Inc. Very large scale immobilized polymer synthesis
US5094594A (en) * 1990-04-23 1992-03-10 Genomyx, Incorporated Piezoelectric pumping device
US5378638A (en) * 1990-08-02 1995-01-03 Boehringer Mannheim Gmbh Analysis element and process for its manufacture
US5491224A (en) * 1990-09-20 1996-02-13 Bittner; Michael L. Direct label transaminated DNA probe compositions for chromosome identification and methods for their manufacture
US5384262A (en) * 1990-11-30 1995-01-24 Quantix Systems, L.P. Quantitative immunoassay for volatile organic compounds
US20040067521A1 (en) * 1990-12-06 2004-04-08 Affymetrix, Inc. Arrays for detecting nucleic acids
US6544739B1 (en) * 1990-12-06 2003-04-08 Affymetrix, Inc. Method for marking samples
US20040029115A9 (en) * 1990-12-06 2004-02-12 Affymax Technologies, N.V. Sequencing of surface immobilized polymers utilizing microfluorescence detection
US5491570A (en) * 1991-07-26 1996-02-13 Accuwave Corporation Methods and devices for using photorefractive materials at infrared wavelengths
US5291763A (en) * 1991-09-18 1994-03-08 Cuisinot Charles P Universal lock for quick release mechanism
US5498530A (en) * 1991-10-16 1996-03-12 Affymax Technologies, N.V. Peptide library and screening method
US5733731A (en) * 1991-10-16 1998-03-31 Affymax Technologies N.V. Peptide library and screening method
US5885837A (en) * 1991-11-22 1999-03-23 Affymetrix, Inc. Very large scale immobilized polymer synthesis using mechanically directed flow paths
US6864101B1 (en) * 1991-11-22 2005-03-08 Affymetrix, Inc. Combinatorial strategies for polymer synthesis
US6849462B1 (en) * 1991-11-22 2005-02-01 Affymetrix, Inc. Combinatorial strategies for polymer synthesis
US6040193A (en) * 1991-11-22 2000-03-21 Affymetrix, Inc. Combinatorial strategies for polymer synthesis
US5484908A (en) * 1991-11-26 1996-01-16 Gilead Sciences, Inc. Oligonucleotides containing 5-propynyl pyrimidines
US5380489A (en) * 1992-02-18 1995-01-10 Eastman Kodak Company Element and method for nucleic acid amplification and detection using adhered probes
US5728802A (en) * 1992-05-06 1998-03-17 Affymax Technologies N.V. Peptides and compounds that bind selectins including endothelium leukocyte adhesion molecule 1 (ELAM-1)
US5607691A (en) * 1992-06-12 1997-03-04 Affymax Technologies N.V. Compositions and methods for enhanced drug delivery
US5288514A (en) * 1992-09-14 1994-02-22 The Regents Of The University Of California Solid phase and combinatorial synthesis of benzodiazepine compounds on a solid support
US5491074A (en) * 1993-04-01 1996-02-13 Affymax Technologies Nv Association peptides
US6864048B2 (en) * 1993-04-28 2005-03-08 Affymetrix, Inc. Factorial chemical libraries
US6852488B2 (en) * 1993-06-25 2005-02-08 Affymetrix, Inc. Identifying a base in a nucleic acid
US5874239A (en) * 1993-07-30 1999-02-23 Affymax Technologies N.V. Biotinylation of proteins
US5723584A (en) * 1993-07-30 1998-03-03 Affymax Technologies N.V. Biotinylation of proteins
US5382512A (en) * 1993-08-23 1995-01-17 Chiron Corporation Assay device with captured particle reagent
US5503805A (en) * 1993-11-02 1996-04-02 Affymax Technologies N.V. Apparatus and method for parallel coupling reactions
US5861476A (en) * 1994-02-02 1999-01-19 Affymax Technologies N.V. Peptides and compounds that bind to the IL-1 receptor
US5608035A (en) * 1994-02-02 1997-03-04 Affymax Technologies N.V. Peptides and compounds that bind to the IL-1 receptor
US5880096A (en) * 1994-02-02 1999-03-09 Affymax Technologies N.V. Peptides and compounds that bind to the IL-1 receptor
US5605793A (en) * 1994-02-17 1997-02-25 Affymax Technologies N.V. Methods for in vitro recombination
US7015046B2 (en) * 1995-03-10 2006-03-21 Mesoscale Technologies, Llc. Multi-array, multi-specific electrochemiluminescence testing
US6368874B1 (en) * 1995-12-22 2002-04-09 Affymax, Inc. Methods for hard-tagging an encoded synthetic library
US5896047A (en) * 1997-02-05 1999-04-20 Xilinx, Inc. Balanced truth-and-complement circuit
US6171797B1 (en) * 1999-10-20 2001-01-09 Agilent Technologies Inc. Methods of making polymeric arrays

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020192684A1 (en) * 1989-06-07 2002-12-19 Affymetrix, Inc. Arrays for detecting nucleic acids
US20050009014A9 (en) * 1989-06-07 2005-01-13 Affymetrix, Inc. Arrays for detecting nucleic acids
US20050158743A1 (en) * 1990-12-06 2005-07-21 Affymetrix, Inc. Arrays for detecting nucleic acids
US20080286786A1 (en) * 2003-11-10 2008-11-20 Investigen, Inc. Methods of preparing nucleic acid for detection

Also Published As

Publication number Publication date
DE69132843T2 (en) 2002-09-12
HK1010219A1 (en) 1999-06-17
EP0562047A1 (en) 1993-09-29
DK0834576T3 (en) 2002-04-22
US20050053928A9 (en) 2005-03-10
EP0834576B1 (en) 2002-01-16
EP0562047A4 (en) 1995-11-02
US20040067521A1 (en) 2004-04-08
US20050170340A9 (en) 2005-08-04
EP0834575A2 (en) 1998-04-08
EP0834575A3 (en) 1999-10-20
DK0834575T3 (en) 2002-04-02
US20030104411A1 (en) 2003-06-05
US20020192684A1 (en) 2002-12-19
US20020155492A1 (en) 2002-10-24
EP0834576A2 (en) 1998-04-08
HK1010218A1 (en) 1999-06-17
EP0834575B1 (en) 2001-11-28
WO1992010588A1 (en) 1992-06-25
AU1248292A (en) 1992-07-08
DE69132905T2 (en) 2002-08-01
US6451536B1 (en) 2002-09-17
US6576424B2 (en) 2003-06-10
US20050112676A1 (en) 2005-05-26
EP1231282A3 (en) 2005-05-18
US20050009014A9 (en) 2005-01-13
US20050164249A1 (en) 2005-07-28
US20050158743A1 (en) 2005-07-21
US20030003475A1 (en) 2003-01-02
US6544739B1 (en) 2003-04-08
US20040248147A1 (en) 2004-12-09
DE69132905D1 (en) 2002-02-21
US20030119011A1 (en) 2003-06-26
DE69132843D1 (en) 2002-01-10
US20030017484A1 (en) 2003-01-23
EP1231282A2 (en) 2002-08-14
US20020164590A1 (en) 2002-11-07
EP0834576A3 (en) 1999-06-16

Similar Documents

Publication Publication Date Title
US6544739B1 (en) Method for marking samples
US6355432B1 (en) Products for detecting nucleic acids
US5925525A (en) Method of identifying nucleotide differences
US5871928A (en) Methods for nucleic acid analysis
US6416952B1 (en) Photolithographic and other means for manufacturing arrays
US6919211B1 (en) Polypeptide arrays
US6551784B2 (en) Method of comparing nucleic acid sequences
US6309822B1 (en) Method for comparing copy number of nucleic acid sequences
US6261776B1 (en) Nucleic acid arrays
US20060194258A1 (en) Polypeptide array synthesis

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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