WO1997043611A1 - Systems and methods for detection of labeled materials - Google Patents

Systems and methods for detection of labeled materials Download PDF

Info

Publication number
WO1997043611A1
WO1997043611A1 PCT/US1997/008319 US9708319W WO9743611A1 WO 1997043611 A1 WO1997043611 A1 WO 1997043611A1 US 9708319 W US9708319 W US 9708319W WO 9743611 A1 WO9743611 A1 WO 9743611A1
Authority
WO
WIPO (PCT)
Prior art keywords
substrate
excitation radiation
focusing
radiation
spot
Prior art date
Application number
PCT/US1997/008319
Other languages
French (fr)
Inventor
David Stern
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
Application filed by Affymetrix, Inc. filed Critical Affymetrix, Inc.
Priority to EP97925604A priority Critical patent/EP0902885A4/en
Priority to JP09541141A priority patent/JP2000512744A/en
Publication of WO1997043611A1 publication Critical patent/WO1997043611A1/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N21/00Investigating or analysing materials by the use of optical means, i.e. using sub-millimetre waves, infrared, visible or ultraviolet light
    • G01N21/62Systems in which the material investigated is excited whereby it emits light or causes a change in wavelength of the incident light
    • G01N21/63Systems in which the material investigated is excited whereby it emits light or causes a change in wavelength of the incident light optically excited
    • G01N21/64Fluorescence; Phosphorescence
    • G01N21/645Specially adapted constructive features of fluorimeters
    • G01N21/6452Individual samples arranged in a regular 2D-array, e.g. multiwell plates
    • 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
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • 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
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N21/00Investigating or analysing materials by the use of optical means, i.e. using sub-millimetre waves, infrared, visible or ultraviolet light
    • G01N21/62Systems in which the material investigated is excited whereby it emits light or causes a change in wavelength of the incident light
    • G01N21/63Systems in which the material investigated is excited whereby it emits light or causes a change in wavelength of the incident light optically excited
    • G01N21/64Fluorescence; Phosphorescence
    • G01N21/6428Measuring fluorescence of fluorescent products of reactions or of fluorochrome labelled reactive substances, e.g. measuring quenching effects, using measuring "optrodes"
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N21/00Investigating or analysing materials by the use of optical means, i.e. using sub-millimetre waves, infrared, visible or ultraviolet light
    • G01N21/62Systems in which the material investigated is excited whereby it emits light or causes a change in wavelength of the incident light
    • G01N21/63Systems in which the material investigated is excited whereby it emits light or causes a change in wavelength of the incident light optically excited
    • G01N21/64Fluorescence; Phosphorescence
    • G01N21/645Specially adapted constructive features of fluorimeters
    • G01N21/6456Spatial resolved fluorescence measurements; Imaging
    • G01N21/6458Fluorescence microscopy
    • GPHYSICS
    • G02OPTICS
    • G02BOPTICAL ELEMENTS, SYSTEMS OR APPARATUS
    • G02B21/00Microscopes
    • G02B21/0004Microscopes specially adapted for specific applications
    • G02B21/002Scanning microscopes
    • G02B21/0024Confocal scanning microscopes (CSOMs) or confocal "macroscopes"; Accessories which are not restricted to use with CSOMs, e.g. sample holders
    • G02B21/0036Scanning details, e.g. scanning stages
    • G02B21/0048Scanning details, e.g. scanning stages scanning mirrors, e.g. rotating or galvanomirrors, MEMS mirrors
    • GPHYSICS
    • G02OPTICS
    • G02BOPTICAL ELEMENTS, SYSTEMS OR APPARATUS
    • G02B21/00Microscopes
    • G02B21/0004Microscopes specially adapted for specific applications
    • G02B21/002Scanning microscopes
    • G02B21/0024Confocal scanning microscopes (CSOMs) or confocal "macroscopes"; Accessories which are not restricted to use with CSOMs, e.g. sample holders
    • G02B21/0052Optical details of the image generation
    • G02B21/0072Optical details of the image generation details concerning resolution or correction, including general design of CSOM objectives
    • GPHYSICS
    • G02OPTICS
    • G02BOPTICAL ELEMENTS, SYSTEMS OR APPARATUS
    • G02B21/00Microscopes
    • G02B21/0004Microscopes specially adapted for specific applications
    • G02B21/002Scanning microscopes
    • G02B21/0024Confocal scanning microscopes (CSOMs) or confocal "macroscopes"; Accessories which are not restricted to use with CSOMs, e.g. sample holders
    • G02B21/0052Optical details of the image generation
    • G02B21/0076Optical details of the image generation arrangements using fluorescence or luminescence
    • GPHYSICS
    • G02OPTICS
    • G02BOPTICAL ELEMENTS, SYSTEMS OR APPARATUS
    • G02B21/00Microscopes
    • G02B21/0004Microscopes specially adapted for specific applications
    • G02B21/002Scanning microscopes
    • G02B21/0024Confocal scanning microscopes (CSOMs) or confocal "macroscopes"; Accessories which are not restricted to use with CSOMs, e.g. sample holders
    • G02B21/008Details of detection or image processing, including general computer control
    • G02B21/0084Details of detection or image processing, including general computer control time-scale detection, e.g. strobed, ultra-fast, heterodyne detection
    • 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
    • B01J2219/00583Features 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
    • B01J2219/00621Delimitation of the attachment areas by physical means, e.g. trenches, raised areas
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00603Making arrays on substantially continuous surfaces
    • B01J2219/00605Making arrays on substantially continuous surfaces the compounds being directly bound or immobilised to solid supports
    • B01J2219/00623Immobilisation or binding
    • B01J2219/00626Covalent
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00603Making arrays on substantially continuous surfaces
    • B01J2219/00605Making arrays on substantially continuous surfaces the compounds being directly bound or immobilised to solid supports
    • B01J2219/00632Introduction of reactive groups to the surface
    • B01J2219/00637Introduction of reactive groups to the surface by coating it with another layer
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00603Making arrays on substantially continuous surfaces
    • B01J2219/00659Two-dimensional arrays

Definitions

  • the present invention generally relates to the field of imaging.
  • the present invention provides scanning systems and methods for high speed imaging of a sample containing labeled materials, and particularly for scanning arrays of polymer sequences, e.g. , oligonucleotide arrays.
  • Confocal microscopes generally employ a pinhole that is confocal with the illuminated spot on the specimen to reject light that is not reflected or emitted from objects in the focal plane. This rejection of out-of-focus light enables the microscope to collect and combine a series of optical slices at different focus positions to generate a two or three dimensional representation of the specimen.
  • Some scanning microscopes employ radiation direction systems, such as galvanometers that include servo-mounted mirrors, to rapidly scan a laser spot across a substrate.
  • these microscopes have relatively high scan rates (e.g., on the order of about 30 lines/second or greater), they generally do not achieve both the resolution and field of view that is necessary for some applications, such as imaging an array of sequenced materials on a substrate.
  • a galvanometer-based confocal microscope's field of view is generally proportional to its resolution. For example, a typical 4Ox microscope objective, which has a 0.25 ⁇ m resolution, has a field size of only about 500 ⁇ m.
  • conventional galvanometer-based confocal microscopes are inadequate for applications requiring both high resolution and a large field of view.
  • Scanning confocal microscope systems such as those discussed in U.S. Patent No. 5,143,854 (Pirrung et al.), PCT WO 92/10092, and U.S. Patent Application Serial No. 08/195,889 (Attorney Docket Number 16528X-006000) , incorporated herein by reference for all purposes, are also known.
  • These scanning systems include an optical train which directs a monochromatic or polychromatic light source to about a 5 micron ( ⁇ m) diameter spot at its focal plane.
  • a photon counter detects the emission from the device in response to the light.
  • the data collected by the photon counter represents one pixel or data point of the image. Thereafter, the light scans another pixel as a translation stage moves the device to a subsequent position.
  • these scanning confocal microscope systems provide high resolution by using an appropriate objective lens and large field of view by using appropriate translation stages.
  • These translation stage-based confocal microscopes obtain high resolution and field of view by sacrificing system throughput.
  • an array of sequenced material using the pioneering fabrication techniques such as those disclosed in U.S. Patent No. 5,143,854 (Pirrung et al.) and U.S. Patent Application Serial No. 08/143,312, incorporated herein by reference for all purposes, may have a density of about 10 5 sequences. Assuming that 36 pixels are required for each sequence, the image can take over at least 10 minutes to acquire.
  • the present invention provides systems, methods and apparatus for detecting marked regions on substrate surfaces.
  • the present invention provides methods and apparatus for scanning a substrate to obtain an image with high sensitivity and resolution at a high speed.
  • the confocal scanning microscopes of the present invention combine the high scan rate of galvanometer based scanning microscopes with a sufficiently high resolution, sensitivity and a large enough field of view for imaging high density arrays of materials, such as those found in the fields of combinatorial chemistry and genetic analysis.
  • the present invention provides a system for detecting marked regions on a surface of a substrate, which comprises an excitation radiation source, and focusing optics for focusing the excitation radiation to regions on the surface of the substrate.
  • a radiation direction system is also included for linearly scanning the focused excitation radiation across the surface of the substrate.
  • a detector is positioned for detecting an emission from the substrate surface in response to the excitation radiation, and a data acquisition system records the amount of detected emission as a function of a position on the surface of the substrate from which the emission was detected.
  • the focusing optics comprises an objective lens having a ratio of scanning field diameter to focused spot diameter of greater than 2000, preferably greater than 3000 and more preferably greater than 4000.
  • the microscope is, for example, capable of focusing a laser beam to a spot having a diameter of about 3 microns at any point within a flat field having a length of about 14 mm.
  • the objective lens has at least a 0.2 numerical aperture, preferably a 0.25 numerical aperture, which provides sufficient sensitivity to detect fluorescently marked regions on the substrate.
  • the radiation direction system preferably comprises a galvanometer mirror that scans the excitation radiation across the surface of the substrate.
  • the objective lens has an external entrance pupil preferably located at or near the galvanometer mirror's pivot location.
  • the mirror usually oscillates at a frequency of at least 7.5 Hz, preferably at least 20 Hz and more preferably at least 30 Hz. In this manner, the laser spot can usually be scanned across the substrate at velocities of about 5 image lines/second, preferably at least 10 images lines/second, and more preferably at least about 30 image lines/second. This allows the microscope to rapidly scan high density substrates, such as the polymer array substrates disclosed by Pirrung. It should be noted that the mirror may scan unidirectionally
  • the galvanometer frequency would generally be about half of the data aquisition speed in image lines/second. Accordingly, the frequency of the galvanometer in the latter case may be lower than 7.5 Hz in order to scan 5 image lines/second.
  • the present invention also provides a system for detecting fluorescent regions on a surface of a substrate, which comprises an excitation radiation source, and first focusing optics for focusing the excitation radiation on the surface of the substrate in a focused spot having a diameter no greater than 10 ⁇ m, preferably less than 5 ⁇ m and more preferably about 3 ⁇ m.
  • An oscillating or reciprocating radiation direction system scans the spot linearly across the surface of the substrate, with a focused travel distance of at least 10 mm and preferably about 14 mm.
  • an optical train separates fluorescence emitted from the surface of the substrate from the excitation radiation reflected from the surface.
  • An autofocus system may also be included for automatically placing the surface of the substrate in a focal plane of the focusing optics.
  • the invention also provides methods of scanning substrates using the above systems.
  • the invention provides a method of scanning a polymer array having a plurality of different polymer sequences, each of the different polymer sequences being immobilized on a surface of a substrate in a different known location, to identify which polymer sequence on the array is bound by a fluorescent target molecule.
  • the method comprises focusing an excitation radiation source upon the surface of the substrate, and scanning the excitation radiation across the surface of the substrate at a speed of at least 5 image lines/second. Fluorescent emissions are collected from the surface of the substrate in response to the excitation radiation. These fluorescent emissions are recorded as a function of a position on the surface of the substrate. The position on the surface indicates the polymer sequence on the array that is bound by the fluorescent target molecule.
  • Fig. 1 is a schematic illustration of one embodiment of a scanning system according to the present invention.
  • Fig. 2 illustrates one embodiment of a scanning system that includes a flow cell on which a substrate is mounted.
  • Fig. 3 is a schematic illustration of a computer based system for controlling and recording data from a scanning system according to the present invention.
  • Fig. 4 is a schematic illustration of an alternate embodiment of a scanning system according to the present invention.
  • Fig. 5 is an enlarged view of a scanned image generated using a scanning system of the present invention.
  • Complementary refers to the topological compatibility or matching together of interacting surfaces of a probe molecule and its target.
  • the target and its probe can be described as complementary, and furthermore, the contact surface characteristics are complementary to each other.
  • Probe is a surface-immobilized molecule that is recognized by a particular target.
  • probes that can be investigated by this invention include, but are not restricted to, agonists and antagonists for cell membrane receptors, toxins and venoms, viral epitopes, hormones (e.g. , opioid peptides, steroids, etc.), hormone receptors, peptides, enzymes, enzyme substrates, cofactors, drugs, lectins, sugars, oligonucleotides, nucleic acids, oligosaccharides, proteins, and monoclonal antibodies.
  • hormones e.g. , opioid peptides, steroids, etc.
  • hormone receptors e.g., enzymes, enzyme substrates, cofactors, drugs, lectins, sugars, oligonucleotides, nucleic acids, oligosaccharides, proteins, and monoclonal antibodies.
  • Target A molecule that has an affinity for a given probe.
  • Targets may be naturally-occurring or manmade molecules. Also, they can be employed in their unaltered state or as aggregates with other species. Targets may be attached, covalently or noncovalently, to a binding member, either directly or via a specific binding substance.
  • targets which can be employed by this invention include, but are not restricted to, antibodies, cell membrane receptors, monoclonal antibodies and antisera reactive with specific antigenic determinants (such as on viruses, cells or other materials), drugs, polynucleotides, nucleic acids, peptides, cofactors, lectins, sugars, polysaccharides, cells, cellular membranes, and organelles. Targets are sometimes referred to in the art as anti-probes. As the term targets is used herein, no difference in meaning is intended.
  • a "Probe Target Pair" is formed when two macromolecules have combined through molecular recognition to form a complex.
  • the present invention provides methods and apparatus for scanning a substrate to obtain a highly sensitive and resolved image at a high speed.
  • the invention will have a wide range of uses, particularly, those requiring quantitative study of a microscopic region from within a larger region, such as 2 or 3 ⁇ m 2 over 100 mm 2 .
  • the invention may find application in the field of histology (for studying histochemical stained and immunological fluorescent stained images) , or fluorescence in situ hybridization.
  • the invention herein is used to image an array of probe sequences fabricated on a support.
  • High resolution scanning systems and methods have routinely been used in the electronics industries, e.g., in the semiconductor and microfabrication industries, to scan microfabricated electronics, e.g., microprocessors, microcircuitry, and the like.
  • microfabricated electronics e.g., microprocessors, microcircuitry, and the like.
  • high resolution scanning methods and devices can be used in the application of polymer arrays. These polymer arrays are generally made up of a large number of different polymer sequences that are coupled to the surface of a typically planar substrate.
  • the substrate on which the polymer sequences are formed may be composed from a wide range of material, either biological, nonbiological, organic, inorganic, or a combination of any of these, existing as particles, strands, precipitates, gels, sheets, tubing, spheres, containers, capillaries, pads, slices, films, plates, slides, etc.
  • the substrate may have any convenient shape, such as a disc, square, sphere, circle, etc.
  • the substrate is preferably flat but may take on a variety of alternative surface configurations.
  • the substrate may contain raised or depressed regions on which a sample is located.
  • the substrate and its surface preferably form a rigid support on which the sample can be formed.
  • the substrate and its surface are also chosen to provide appropriate light-absorbing charac ⁇ teristics.
  • the substrate may be a polymerized Langmuir Blodgett film, functionalized glass, Si, Ge, GaAs, GaP, Si0 2 , SiN 4 , modified silicon, or any one of a wide variety of gels or polymers such as (poly)tetrafluoroethylene, (poly)vinylidenedifluoride, polystyrene, polycarbonate, or combinations thereof.
  • the substrate is flat glass or silica.
  • the surface of the substrate is etched using well known techniques to provide for desired surface features.
  • the synthesis regions may be more closely placed within the focus point of impinging light.
  • the surface may also be provided with reflective "mirror" structures for maximization of emission collected therefrom.
  • the surface will usually, though not always, be composed of the same material as the substrate.
  • the surface may be composed of any of a wide variety of materials, for example, polymers, plastics, resins, polysaccharides, silica or silica-based materials, carbon, metals, inorganic glasses, membranes, or any of the above- listed substrate materials.
  • the surface will be optically transparent and will have surface Si-OH functionalities, such as those found on silica surfaces.
  • arrays are typically prepared in a manner that allows groups of each different polymer sequence to be coupled in a different known location on the substrate surface.
  • the pioneering methods for synthesizing these arrays have been previously described. For example, methods of synthesizing arrays of polymers using light directed synthesis methods are described in U.S. Patent No. 5,143,854 to Pirrung et al., and U.S. Patent No. 5,405,783 to Fodor et al. , the complete disclosures of which are incorporated herein by reference. Additionally, mechanical methods for synthesizing these arrays are described in, e.g., U.S. Patent No. 5,384,261 to Winkler et al., the complete disclosure of which is incorporated herein by reference.
  • the combination of photolithograpic and fabrication techniques may, for example, enable each probe sequence ("feature") to occupy a very small area ("site") on the support.
  • this feature site may be as small as a few microns or even a single molecule.
  • Such probe arrays may be of the type known as Very Large Scale Immobilized Polymer Synthesis (VLSIPSTM) .
  • VLSIPSTM Very Large Scale Immobilized Polymer Synthesis
  • the arrays for use with the present invention are described in terms of arrays of different oligonucleotide sequences. However, it should be readily understood that a variety of different polymer array types are equally applicable to the devices of the invention.
  • the scanners of the present invention may also be used in the reflectance mode to scan non-fluorescent surfaces.
  • such arrays may be prepared having all nucleotide sequences of a given length, composed of the basis set of monomers.
  • an array of oligonucleotides containing all possible sequences of length n which is made up of the basis set of four nucleotides contains up to 4 n oligonucleotides on its surface.
  • such arrays may include upwards of about 65,536 and 1,048,576 different oligonucleotides, respectively.
  • a simple binary masking strategy can be used, as described in U.S. Patent No.
  • Alternate masking strategies can produce arrays of probes which contain a subset of polymer sequences, i.e., polymers having a given subsequence of monomers, but are systematically substituted at each position with each member of the basis set of monomers.
  • these alternate synthesis strategies may be used to lay down or "tile" a range of probes that are complementary to, and span the length of a given known nucleic acid segment.
  • the tiling strategy will also include substitution of one or more individual positions within the sequence of each of the probe groups with each member of the basis set of nucleotides.
  • interrogation positions By reading the hybridization pattern of the target nucleic acid, one can determine if and where any mutations lie in the sequence, and also determine what the specific mutation is by identifying which base is contained within the interrogation position. Tiling methods and strategies are discussed in substantial detail in Published PCT Application No. 95/11995, the complete disclosure of which is incorporated herein by reference in its entirety for all purposes.
  • Tiled arrays may be used for a variety of applications, such as identifying mutations within a known oligonucleotide sequence or "target".
  • the probes on the array will have a sub-sequence which is complementary to a known nucleic acid sequence, but wherein at least one position in that sequence has been systematically substituted with the other three nucleotides. See, e.g., U.S. Patent No. 5,527,681, the complete disclosure of which is incorporated herein by reference.
  • the sample nucleic acid for which sequence information is desired is contacted with the array.
  • This "target" sequence is typically labeled with a detectable group such as a fluorescent moiety, i.e., fluorescein or rhodamine.
  • a detectable group such as a fluorescent moiety, i.e., fluorescein or rhodamine.
  • the position on the array to which the target sequence binds by scanning the surface of the array for fluorescence.
  • the surface is typically scanned by directing excitation radiation at the surface to activate the fluorescent labeling group in the target, which in turn emits a fluorescent response radiation.
  • the fluorescent response radiation is detected and assigned to the region from which it originated. By knowing the position from which the fluorescence originates, one can identify the sequence to which the target binds.
  • the term "feature” generally refers to any element, e.g. , region, structure or the like, on the surface of a substrate.
  • substrates to be scanned using the scanning systems described herein will have small feature sizes, and consequently, high feature densities on substrate surfaces.
  • individual features will typically have at least one of a length or width dimension that is no greater than 100 ⁇ m, and preferably, no greater than 50 ⁇ m, and more preferably greater than about 20 ⁇ m.
  • a length or width dimension that is no greater than 100 ⁇ m, and preferably, no greater than 50 ⁇ m, and more preferably greater than about 20 ⁇ m.
  • each different polymer sequence will typically be substantially contained within a single feature.
  • the probe arrays will have a wide range of applications.
  • the probe arrays may be designed specifically to detect genetic diseases, either from acquired or inherited mutations in an individual DNA. These include genetic diseases such as cystic fibrosis, diabetes, and muscular dystrophy, as well as acquired diseases such as cancer (P53 gene relevant to some cancers) , as disclosed in U.S. Patent Application Serial Number 08/143,312, already incorporated by reference.
  • Genetic mutations may be detected by a method known as sequencing by hybridization.
  • sequencing by hybridization a solution containing one or more targets to be sequenced (i.e., samples from patients) contacts the probe array.
  • the targets will bind or hybridize with complementary probe sequences.
  • the targets are labeled with a fluorescent marker, radioactive isotopes, enzymes, or other types of markers. Accordingly, locations at which targets hybridize with complimentary probes can be identified by locating the markers. Based on the locations where hybridization occur, information regarding the target sequences can be extracted. The existence of a mutation may be determined by comparing the target sequence with the wild type.
  • targets and probes can be characterized in terms of kinetics and thermodynamics. As such, it may be necessary to interrogate the array while in contact with a solution of labeled targets. Consequently, the detection system must be extremely selective, with the capacity to discriminate between surface-bound and solution- born targets. Also, in order to perform a quantitative analysis, the high-density volume of the probe sequences requires the system to have the capacity to distinguish between each feature site.
  • the devices of the present invention generally employ a scanning device which rapidly sweeps an activation radiation beam or spot across the surface of the substrate.
  • the devices also include focusing optics for focusing the excitation radiation onto the surface of the substrate in a sufficiently small area to provide high resolution of features on the substrate, while simultaneously providing a wide scanning field.
  • An image is obtained by detecting the electromagnetic radiation emitted by the labels on the sample when the labels are illuminated.
  • fluorescent emissions are gathered by the focusing optics and detected to generate an image of the fluorescence on the substrate surface.
  • the devices of the invention further employ confocal detection systems to reduce or eliminate unwanted signals from structures above and below the plane of focus of the excitation radiation, as well as autofocus systems to focus both the activation radiation on the substrate surface and the emitted radiation from the surface.
  • the excitation radiation and response emission have different wavelengths. Filters having high transmission in the label's emission band and low transmission in the excitation wavelength may be utilized to inhibit the detection of undesirable emission. These generally include emission from out-of-focus planes or scattered excitation illumination as potential sources of background noise.
  • the devices of the present invention include one or more sources of excitation radiation.
  • these source(s) are immobilized or stationary point light sources, e.g., lasers such as argon, helium-neon, diode, dye, titanium sapphire, frequency-doubled diode pumped Nd:YAG and krypton.
  • the excitation source illuminates the sample with an excitation wavelength that is within the visible spectrum, but other wavelengths (i.e., near ultraviolet or near infrared spectrum) may be used depending on the application (i.e., type of markers and/or sample).
  • the sample is excited with electromagnetic radiation having a wavelength at or near the absorption maximum of the species of label used.
  • Exciting the label at such a wavelength produces the maximum number of photons emitted. For example, if fluorescein (absorption maximum of 488 nm) is used as a label, an excitation radiation having a wavelength of about 488 nm would induce the strongest emission from the labels.
  • a wavelength which approximates the mean of the various candidate labels' absorption maxima may be used.
  • multiple excitations may be performed, each using a wavelength corresponding to the absorption maximum of a specific label. Table I lists examples of various types of fluorophores and their corresponding absorption maxima.
  • the excitation radiation from the point source is directed at a movable radiation direction system which rapidly scans the excitation radiation beam back and forth across the surface of the substrate.
  • a variety of devices may be employed to generate the sweeping motion of the excitation radiation.
  • resonant scanner or rotating polygons may be employed to direct the excitation radiation in this sweeping fashion.
  • galvanometer devices are preferred as the scanning system.
  • the term "galvanometer” refers to devices that employ a servo motor to oscillate or rotate a mirror over a restricted, predefined range, which is typically less than 90°.
  • an optical train may be employed between the activation source and the galvanometer mirror to assist in directing, focusing or filtering the radiation directed at and reflected from the galvanometer mirror.
  • the galvanometers employed in the devices and systems of the present invention typically sweep a scanning spot across the substrate surface at an oscillating frequency that is typically greater than about 7.5 Hz, preferably greater than about 20 Hz and more preferably, greater than about 30 Hz. With this frequency, the spot can typically be scanned across the substrate at a velocity of at least about 20 lines/second, preferably in about 5 image lines/second, preferably at least 10 images lines/second, and more preferably at least about 30 image lines/second. It should be noted that the mirror may scan unidirectionally (e.g. , with a sawtooth wave) or bidirectionally (e.g., with a symmetric triangle wave) .
  • the galvanometer frequency would generally be about half of the data aquisition speed in image lines/second. Accordingly, the frequency of the galvanometer in the latter case may be lower than 7.5 Hz in order to scan 5 image lines/second.
  • the activation radiation is then directed through focusing optics to focus the sweeping beam at the surface of the array which is used to interrogate the target sequence. These same focusing optics also collect emitted fluorescence from the substrate surface for subsequent detection.
  • the objective lens is preferably selected to provide high resolution, as determined by the focused spot size, while still allowing a wide scanning field. "Focused spot size" is defined by the diameter of the focused activation spot on the surface of the substrate at 1/e 2 intensity points.
  • “Scanning field” is defined as the length of travel of the focused activation beam or “spot,” in one dimension parallel to the direction of travel of the sweeping beam.
  • the confocal system of the present invention generally provides a focused spot size having a diameter no greater than about 10 ⁇ m, preferably no greater than about 5 ⁇ m, and more preferably about 3 ⁇ m.
  • these systems have a scanning field with a dimension parallel to the direction of the sweep of the activation beam that is usually greater than about 10 mm and more preferably about 14 mm.
  • the combined measurement of resolution and effective scanning field are provided as a ratio of the size of the effective field of view of the objective to the size of the focused spot from the scanner.
  • a scanner having a scanning field of 2 mm and a focused spot size of 10 ⁇ m would have a ratio of 200 (2mm/0.010 mm). While a scanner having an effective scanning field of 14 mm with a focused spot size of 3 ⁇ m would have a ratio of 4666.
  • the scanning system of the present invention usually has a ratio of size of effective field of view to size of focused spot of at least about 2000, preferably greater than about 3000, and more preferably greater than about 4000.
  • scanning field generally refers to the direction in which the galvanometer mirror scans the laser spot across the substrate. The translation stage moves the substrate in the direction orthogonal to the scanned direction, and this direction may be larger than 14 mm.
  • high fluorescence collection efficiency is achieved by providing an objective lens having a numerical aperture that is at least about 0.2.
  • the numerical aperture is at least about 0.25. This translates to an F/number of about F/2.5 to F/2.
  • the focusing optics of the scanning system may include a telecentric objective lens. This allows the activation beam to strike the scanning surface at an angle that is substantially normal to that surface, across the entire scanning surface, i.e., regardless of where the input beam is propagating through the lens relative to the lens axis.
  • substantially normal is meant an angle of incidence of approximately 0°, e.g., between 0° and 5°, and preferably from 0° to 2°.
  • the activation radiation spot As the activation radiation spot is swept across the surface of the substrate, it activates any fluorescent groups that remain upon the surface, e.g., those that are bound to the surface.
  • the activated groups emit a response radiation or emission which is then collected by the objective lens and directed back through the optical train via the servo mounted mirror.
  • dichroic mirrors or beam splitters may be included in the optical train. These dichroic beam splitters or mirrors are reflective to radiation in the wavelength of the excitation radiation while transmissive to radiation in the wavelength of the response radiation. For example, where an Argon laser is used as the point energy source, it will typically generate activation radiation having a wavelength of about 488 nm.
  • Fluorescence emitted from an activated fluorescein moiety on the other hand will typically have a wavelength between about 515 and 545 nm.
  • dichroic mirrors may be included which transmit light having a wavelength greater than 515 nm while reflecting light of shorter wavelengths. This effectively separates the excitation radiation reflected from the surface of the substrate from the response radiation emitted from the surface of the substrate.
  • additional dichroic mirrors may be used to separate signals from label groups having different response radiation wavelengths, thereby allowing simultaneous detection of multiple fluorescent indicators, and thus, simultaneous interrogation of a single array with multiple target sequences.
  • a first target sequence may be labeled with a shorter wavelength fluorescent label, e.g., fluorescein, while a second target sequence is labeled with a longer wavelength fluorescent label, e.g., carbocyanin dyes such as CY3 which emit response radiation in the range of 550 to 600 nm.
  • the response radiation from each of these targets may be separated and individually detected through the inclusion of additional dichroic beam splitters and detectors. Following separation of the response radiation from the reflected excitation radiation, the response radiation or fluorescence is then directed at a detector, e.g., a photomultiplier tube, to measure the level of response radiation and record that level as a function of the position on the substrate from which that radiation originated.
  • a detector e.g., a photomultiplier tube
  • the response radiation is focused upon the detector through a spatial filter such as a confocal pinhole.
  • a spatial filter such as a confocal pinhole.
  • the device may incorporate a bandpass filter between the dichroic mirror and the detector to further restrict the wavelength of radiation that is delivered to the detector.
  • the reflected excitation radiation may generally be used in an autofocusing system in the devices described herein.
  • the reflected excitation radiation may also be directed at a detector, e.g., a photodiode, that is preferably located behind another spatial filter, i.e., a confocal pinhole.
  • the substrate surface may be moved closer to and further from the objective lens using, e.g., a translation stage upon which the substrate is mounted.
  • a translation stage upon which the substrate is mounted.
  • the amount of reflected excitation radiation contacting the photodiode will be reduced.
  • this amount will increase, reaching a maximal value when the substrate is in focus.
  • Control for the autofocus system is generally supplied by an appropriately programmed computer, which moves the translation stage in response to the input from the photodiode, until a maximum is reached.
  • this computer is the same computer which receives, compiles and stores the input from the fluorescence detector or photomultiplier tube, to produce an output of fluorescence as a function of position on the array, typically in the form of a numerical representation or a scan image.
  • Examples of autofocus confocal systems for use in fluorescent scanning devices are generally described in commonly assigned, co- pending U.S. Patent Application Serial No. 08/195,889, filed February 10, 1994, the complete disclosure of which is incorporated herein by reference for all purposes.
  • Fig. 1 is an optical block diagram illustrating the imaging system 100 according to the present invention.
  • imaging system 100 is used to obtain images of oligonucleotide probe arrays to which fluorescently labelled DNA or RNA is bound. It might also be used for other applications, such as wafer or mask inspection, or for imaging of polypeptide or other polymer arrays, electrophoresis gels, or biological specimens.
  • a beam of excitation radiation e.g., 488-nm light
  • a photodetector 131 optionally a photodiode used as a laser power monitor.
  • the portion of the beam that is transmitted through beamsplitter 104 is reflected by dichroic beamsplitters 106 and 108 and transmitted through lenses 111 and 112, which provide a telescope to expand the beam emitted by laser 102 and to demagnify the collimated fluorescence obtained from the surface of the substrate.
  • lens ill has a focal length of 20 mm and lens 112 has a focal length of 80 mm. Other focal lengths and focal length ratios can be used, but system performance (confocality, resolution, etc.) may be affected.
  • the expanded laser beam is reflected by mirror 114 and focused by lens 116, which is described in more detail below.
  • Mirror 114 which typically is an 18 mm x 28 mm octagon or ellipse, is a galvanometer mirror that oscillates (i.e., scans) at a rate of typically several Hertz or several tens of Hertz. In preferred aspects, the galvanometer mirror oscillates at frequencies greater than 7.5 Hz, preferably about 20 Hz and more preferably about 30 Hz.
  • a resonant scanner or rotating polygonal mirror could be used instead of a galvanometer.
  • imaging system 100 obtains an image of a 2-dimensional area by scanning the laser beam in one dimension while moving the sample in an orthogonal direction.
  • sample 118 is positioned such that the plane of interest (e.g.
  • Remitted light is collected by lens 116, reflected by mirror 114, and transmitted through lenses 112 and 111.
  • Remitted light may consist of fluorescence, phosphorescence, specular reflection, diffuse reflection, Raman scattering, etc.
  • Remitted light having a wavelength greater than 555 nm passes through dichroic beamsplitter 108 and is focused by lens 134 onto pinhole 136.
  • Remitted light having a wavelength between 515 and 555 nm is reflected by beamsplitter 108, passes through dichroic beamsplitter 106, and is focused by a lens 120 onto a pinhole 122.
  • Filter 138 transmits light having wavelengths between 555 and 607 nm
  • filter 124 transmits light having wavelengths between 515 and 545 nm.
  • Remitted light having wavelengths less that 515 nm is reflected by beamsplitters 108 and 106 and partially reflected by beamsplitter 104; the light reflected by beamsplitter 104 is focused by a lens 132 onto a pinhole 128.
  • the light that is transmitted through pinhole 128 impinges upon a photodetector 130.
  • Pinholes 136, 122, and 128 are typically confocal pinholes. Specifically, remitted light that originates from the location on sample 118 where the laser beam is focused is maximally transmitted through the pinholes, while light that originates from other locations is not.
  • Lenses 134, 120, and 132 preferably have focal lengths of 50 mm.
  • Pinholes 136 and 122 preferably have diameters of 100 microns; pinhole 128 preferably has a diameter of 50 microns. Other lens focal lengths and pinhole diameters can be used, but system performance (confocality, sensitivity to misalignment, etc.) may be affected.
  • pinholes 136 and 122 If lens 116 is not fully corrected for lateral color, the remitted light that is focused onto pinholes 136 and 122 might move laterally (across the plane of the pinhole) as mirror 114 is scanned. In this case it might be advantageous to use pinholes that are elliptical or rectangular rather than round, or to use slits instead of pinholes.
  • lens 116 in the present scanner has about 3 microns of lateral color at the edges of the scan field; consequently, the remitted light can move laterally by about 40 microns across the pinholes.
  • Reasonable sizes for pinholes 136 and 122 in this case might be approximately 75 microns by 125 microns.
  • Photodetectors 140 and 126 are typically photomultipliers and are typically intended for the detection of relatively weak signals such as fluorescence.
  • Various light detectors may be used, including photodiodes, avalanche photodiodes, phototransistors, vacuum photodiodes, photomultiplier tubes, and other light detectors.
  • photodetector 130 is used to aid in focusing the sample prior to scanning or while it is being scanned.
  • the amount of reflected laser light reaching photodetector 130 is a maximum when the laser beam is focused onto the surface of sample 118. Focusing can be done either manually, or by a motorized translation stage under computer control.
  • sample 118 is a glass slide mounted in a liquid-filled flow cell (see Fig. 2) and the surface of interest is the second surface of the glass, i.e. the glass- liquid interface.
  • the reflection from this surface is much weaker than the reflection from the first surface of the glass, i.e. the glass-air interface, but the system is sufficiently confocal that the reflection peaks from the 2 surfaces are well separated.
  • the system described above is particularly useful in the detection of fluorescence from targets that are labelled with dyes such as fluorescein and phycoerythrin.
  • dyes such as fluorescein and phycoerythrin.
  • Other dyes rhodamines, carbocyanines, etc.
  • laser 102, beamsplitters 108 and 106, and filters 138 and 124 may need to be altered depending on the dyes' absorption and emission spectra.
  • target molecules may be labeled with particles or very large molecules that scatter laser light but do not fluoresce. In these cases, it may be desirable to detect diffusely scattered light but not specularly reflected laser light.
  • An annular aperture can be placed between beamsplitter 104 and lens 132. If the inner diameter of the annulus is selected properly, this aperture will block specularly reflected laser light while transmitting diffusely scattered laser light, which can then be detected by photodetector 130.
  • the objective lens 116 was designed to meet a number of specifications, and may be obtained from, e.g. , Special Optics, Part No. 55-S30-15(Wharton, NJ) .
  • the objective lens is usable with a galvanometer (or resonant scanner, rotating polygon, etc.), i.e. with an entrance pupil that is external to the lens and is located at or near the galvanometer mirror's pivot location.
  • the lens is also capable of focusing a TEM 00 -mode laser beam having a wavelength of 488 nm to a spot having a diameter no greater than 3 microns at the 1/e 2 intensity points, and should be capable of doing this at any point within a flat field having a diameter no less than 14 mm.
  • Objective lens 116 is within 2 degrees of telecentric, i.e. the transmitted laser beam should be parallel to the optical axis of the lens to within 2 degrees. (If the lens is not telecentric or nearly so, specularly reflected light from near the edges of the field might miss the lens and therefore not be detected.) In addition, lens 116 was designed so that the F/number would be no greater than f/2, i.e. numerical aperture no less than 0.25, with no vignetting even at the edges of the field. (F/number is a measure of the lens's light-gathering ability and is important if the lens is to be used for the detection of weak signals) .
  • Objective lens 116 is capable of focusing a polychromatic beam, having wavelengths between 488 and 600 nm and filling the entrance pupil, to a spot having a diameter no greater than 10 microns, and is capable of doing this at any point within a flat field having a diameter no less than 14 mm.
  • Objective lens 116 preferably has a focal length of about 30 mm. A lens having a different focal length, but still meeting the above specifications could be readily designed and would be acceptable for the purposes described herein.
  • Lens 116 can focus a 488 nm laser beam to a diffraction-limited spot when the ratio of input beam diameter to lens focal length is less than approximately 0.21, e.g. it can focus a 6.2 mm diameter beam to a 3 micron diameter spot. If a larger focal spot is desired, the input beam diameter can be made smaller, e.g. by changing the ratio of the focal lengths of lenses 111 and 112 or by demagnifying the beam between laser 102 and beamsplitter 104.
  • Lens 116 was designed to be able to image a 14 mm field with 3 micron resolution and 0.25 numerical aperture; however, the design can easily be scaled up, e.g. to image a 28 mm field with 6 micron resolution and 0.25 numerical aperture (or potentially, to image a 280 mm field with 60 micron resolution and 0.25 numerical aperture, although the lens would be extremely large and costly) .
  • This scale-up involves merely multiplying the thicknesses, diameters, and radii of curvature of all of the elements of lens 116 by a constant.
  • the entire scanner can be scaled up for imaging of larger areas by multiplying all of the lens diameters and focal lengths, pinhole diameters, and mirror diameters by the same factor.
  • imaging system 300 includes a galvanometer 306 (to which a mirror 114 is attached) and a galvanometer driver board 305, which can be obtained from General Scanning Inc. (Watertown, MA) .
  • Galvanometer 306 is a model M2T. Other suitable galvanometers and driver boards are available, e.g. from Cambridge Technology Inc. (Watertown, MA) .
  • the input to driver board 305 is a voltage waveform from arbitrary waveform generator 304 (Keithley Metrabyte model PCIP-AWFG, Taunton, MA) , which is installed in an ISA slot in computer 302.
  • Circuitry on board 305 attempts at every instant to force galvanometer 306 to whatever angular position is commanded by waveform generator 304 (the desired angular position is linearly related to the waveform voltage) .
  • Waveform generator 304 after being programmed, can generate a waveform indefinitely without further intervention by computer 302.
  • the waveform used is typically a sawtooth wave: e.g. if the waveform period is 33.3 msec, the voltage ramps up linearly for 25 msec, during which time data are acquired as the laser beam sweeps across the field; during the next 8.3 msec the voltage returns to its initial value and the laser beam retraces.
  • Other waveforms such as sine waves or symmetric triangle waves could be used.
  • Filter cutoff frequencies are set by software so that each filter's output rise time in response to a step input is approximately equal to the time between A/D conversions. For example, if A/D conversions are 6 microseconds apart, the cutoff frequencies are set to 66 kHz.
  • filters filters with more or fewer poles of different transfer functions (e.g. Butterworth) , fixed-frequency filters, or simple RC filters) could be used instead.
  • Filter outputs are digitized by a 12-bit, 330-kHz analog-to digital converter on data acquisition board 308 (Computer Boards Inc. model CIO-DAS16/330) . Similar data acquisition boards are available from several other manufacturers. Digitized data are displayed by computer 302, typically in the form of a gray-scale image, and are written to the computer's hard disk for subsequent data analysis. If the internal clock on data acquisition board 308 is used to trigger A/D conversions, the images obtained may be slightly distorted.
  • A/D conversions that are equally far apart in time are not equally far apart in space, i.e., the velocity of the laser beam across sample 118 is not exactly constant, partly because lens 116 is not an ideal f-theta lens, and partly because of non-ideal behavior by driver board 305 and galvanometer 306. Therefore, A/D conversions are preferably triggered by waveform generator 304, which has digital output channels in addition to its analog output channel. When waveform generator 304 is appropriately programmed and its digital output pulses are appropriately spaced, image distortion can be eliminated.
  • A/D conversions are performed per scan line, and if the user wants fewer than 4096 pixels/line the A/D conversions are binned by software in groups of 2 or more. For example, if the length of a scan line is 14 mm, the laser beam moves 3.4 microns between one A/D conversion and the next; and if, for example, the user chooses a pixel size of 13.6 microns, software adds the numbers from 4 A/D conversions for each pixel.
  • One version of the software allows 8192 A/D conversions per scan line. The utility of doing more A/D conversions per line diminishes as the pixel size becomes less than the laser spot size.
  • Translation stages 332 are controlled by indexer 330.
  • Two axes of translation are usually used: a scan axis (parallel to the plane of the sample and perpendicular to the direction of motion of the laser spot) , and a focus axis (parallel to the optical axis of lens 116) .
  • a third axis orthogonal to the other two may also be desirable.
  • Computer 302 sends commands to indexer 330, and receives information about translation- stage status from indexer 330, e.g., using an RS-232 interface.
  • Translation stage speed is set such that the distance that sample 118 moves per scan line is equal to the desired pixel size.
  • Software allows the user to control various scan parameters, including the size of the area to be scanned, the pixel size, the scan speed, and which output or outputs to digitize (either of the photomultiplier outputs, or the photodiode output, or a combination thereof) .
  • Default parameters are 14 mm x 14 mm scan area, 10.2 micron pixel size, and scan speed of 30 lines/second. With these parameters, an image contains 1365 x 1365 pixels and can be acquired in about 45 seconds.
  • an image of a 14 mm x 14 mm area contains 4096 x 4096 pixels and can be acquired in about 2 minutes and 16 seconds. If an image with adequate signal-to noise ratio is not obtained at a scan speed of 30 lines/second, the user might choose to reduce the scan speed, for example to 7.5 lines/second (quadrupling the scan time approximately quadruples the number of photons detected per pixel, and therefore in many cases approximately doubles the signal-to-noise ratio) .
  • Scanned areas can vary from the 14 mm x 14 mm dimensions described above, and need not be square.
  • software options can allow the user to reduce the length of a scan line, e.g., to as little as 4 mm or smaller if a user so desired, with a pixel size as small as 1 micron, and to set the number of lines per scan independently of the other scan parameters.
  • a single scan can not cover a region that is larger than the field of view of lens 116, e.g., 14 mm.
  • the system may cover a region that is several inches wide in the orthogonal direction (limited only by the length of travel of the translation stage) .
  • sample 118 may be part of a packaged chip, such as a diced chip glued into a disposable plastic package.
  • a packaged chip such as a diced chip glued into a disposable plastic package.
  • imaging system 100 further includes a body
  • the support may be a microscope slide or any surface which is adequate to hold the sample.
  • the body 422, depending on the application, may be a flow cell having a cavity 423.
  • the flow cell for example, may be employed to detect reactions between targets and probes.
  • the bottom of the cavity may comprise a light absorptive material so as to minimize the scattering of incident light.
  • surface 431 is mated to body 422 and serves to seal cavity 423.
  • the flow cell and the substrate may be mated for sealing with one or more gaskets.
  • the substrate is mated to the body by vacuum pressure generated by a pump 452.
  • the flow cell is provided with two concentric gaskets and the intervening space is held at a vacuum to ensure mating of the substrate to the gaskets.
  • the substrate may be attached by using screws, clips, or other mounting techniques.
  • the cavity encompasses the sample.
  • the cavity includes an inlet port 421 and an outlet port 420.
  • a fluid which in some embodiments contains fluorescently labeled targets, is introduced into the cavity through inlet port 421.
  • a pump 453 which may be a model no. B-120-S made by Eldex Laboratories, circulates fluids into the cavity via inlet 421 port and out through outlet port 420 for recirculation or disposal.
  • a syringe, gas pressure, or other fluid transfer device may be used to flow fluids into and through the cavity.
  • pump 453 may be replaced by an agitation system that agitates and circulates fluids through the cavity. Agitating the fluids shortens the incubation period between the probes and targets. This can be best explained in terms of kinetics.
  • a thin layer known as the depletion layer, is located above the probe sample. Since targets migrate to the surface and bind with the probe sequences, this layer is essentially devoid of targets. However, additional targets are inhibited from flowing into the depletion layer due to finite diffusion coefficients. As a result, incubation period is significantly increased.
  • the agitation system to dissolve the depletion layer, additional targets are presented at the surface for binding. Ultrasonic radiation and/or heat, shaking the holder, magnetic beads, or other agitating technique may also be employed.
  • the flow cell is provided with a temperature controller 450 for maintaining the flow cell at a desired temperature. Since probe/target interaction is sensitive to temperature, the ability to control it within the flow cell permits hybridization to be conducted under optimal temperature.
  • the temperature controller 450 may include a circulating bath, a refrigerated air circulating device, resistance heater, peltier device (thermoelectric cooler) , or other temperature controller may be implemented.
  • flow cell 422 may mounted to a translation stage 424 for moving the flow cell in an orthogonal direction relative to the optical path.
  • the flow cell may be mated to the translation stage by vacuum pressure generated by pump 452.
  • screws, clips or other mounting techniques may be employed to mate the flow cell to the translation stage.
  • imaging system 400 comprises components which are common to the system described in Fig. 1.
  • lens 116 is intended for use at a finite rather than an infinite conjugate ratio, and therefore lens 112 is omitted.
  • This scanner uses only a single pinhole 401, and all of the remitted light passes through this pinhole 401 before being separated by the various dichroic beamsplitters.
  • This scanner has 2 additional dichroic beamsplitters, spectral filters, and photomultipliers, and therefore can be used for 4-channel rather than merely 2-channel fluorescent scanning.
  • a beam of excitation radiation e.g., 488-nm light
  • a beamsplitter 104 partially reflected and partially transmitted by a beamsplitter 104.
  • the reflected portion of the beam impinges upon a photodetector 131 (optional) , which is typically a photodiode used as a laser power monitor.
  • the portion of the beam that is transmitted through beamsplitter 104 is reflected by dichroic beamsplitter 106 and transmitted through lens 111, focused onto pinhole 401, and expanded to the desired diameter at the entrance pupil lens 116.
  • the beam of excitation radiation is focused by lens
  • imaging system 300 may be constructed such that remitted light having a wavelength between 515 and 545 is reflected by dichroic beamsplitter 410, and passes through a filter 412 onto photodetector 414.
  • Remitted light having a wavelength between 545 and 570 nm may be reflected by dichroic beamsplitter 416, where it passes through filter 418 and onto photodetector 420.
  • beamsplitter 422 is constructed to reflect remitted light having wavelengths between 570 and 595 nm through filter 423 onto photodetector 426. Remitted light having wavelengths greater than 595 nm are transmitted through beamsplitter 422, to pass through filter 425 onto photodetector 424.
  • a tiling array was designed for the entire human mitochondrial genome. See M. Chee et al., "Accessing genetic information with high-density DNA arrays", Science vol. 274, pgs. 610-614 (1996).
  • the array contained 134,688 different probe sequences, each occupying a separate 35 ⁇ m feature. The overall array dimensions were 1.28 cm X 1.28 cm.
  • RNA polymerase promoter tagged primers a 16.3 kb fragment of mtDNA was amplified directly from genomic DNA samples. Labeled 16.3 kb RNA targets were prepared by in vitro transcription from the PCR amplicons and hybridized to the array.
  • a scanning system as described above was used to scan the array and a 4096X4096 pixel image was obtained, each pixel representing 3.4 ⁇ m.
  • the entire array was scanned in under three minutes.
  • a close-up view of a scanned image of this array is shown in Figure 5.
  • the scanned image shown covers a 1.5 mm X 1.5 mm segment of 14 mm X 14 mm image of a 12.8 mm X 12.8 mm array.

Abstract

Labeled targets on a support synthesized with polymer sequences at known locations can be detected by exposing marked regions of sample to radiation from a source (102) and detecting (140, 126) the emission thereform, and repeating the steps of exposition and detection until the sample is completely examined.

Description

SYSTEMSANDMETHODS FOR DETECTIONOF LABELEDMATERIALS
CROS8-REFERENCE TO RELATED APPLICATIONS
The present application is a regular application of provisional U.S. Patent Application Serial No. 60/017,203, filed May 16, 1996 (Attorney Docket No. 16528X-018900) , the complete disclosure of which is hereby incorporated herein by reference for all purposes. This application is also related to commonly assigned, co-pending U.S. Patent Application Serial No. 08/301,051, filed September 9, 1994, now U.S. Patent No. 5,578,832, and U.S. Patent Application Serial No. 08/195,889, filed February 10, 1994 (Attorney Docket Nos.
16528X-003800 and 16528X-006000, respectively) the complete disclosures of which are incorporated herein by reference for all purposes.
BACKGROUND OF THE INVENTION
The present invention generally relates to the field of imaging. In particular, the present invention provides scanning systems and methods for high speed imaging of a sample containing labeled materials, and particularly for scanning arrays of polymer sequences, e.g. , oligonucleotide arrays.
Methods and systems for imaging samples containing labeled markers such as confocal microscopes are commercially available. Confocal microscopes generally employ a pinhole that is confocal with the illuminated spot on the specimen to reject light that is not reflected or emitted from objects in the focal plane. This rejection of out-of-focus light enables the microscope to collect and combine a series of optical slices at different focus positions to generate a two or three dimensional representation of the specimen.
Some scanning microscopes employ radiation direction systems, such as galvanometers that include servo-mounted mirrors, to rapidly scan a laser spot across a substrate. Although these microscopes have relatively high scan rates (e.g., on the order of about 30 lines/second or greater), they generally do not achieve both the resolution and field of view that is necessary for some applications, such as imaging an array of sequenced materials on a substrate. In fact, a galvanometer-based confocal microscope's field of view is generally proportional to its resolution. For example, a typical 4Ox microscope objective, which has a 0.25 μm resolution, has a field size of only about 500 μm. Thus, conventional galvanometer-based confocal microscopes are inadequate for applications requiring both high resolution and a large field of view.
Scanning confocal microscope systems, such as those discussed in U.S. Patent No. 5,143,854 (Pirrung et al.), PCT WO 92/10092, and U.S. Patent Application Serial No. 08/195,889 (Attorney Docket Number 16528X-006000) , incorporated herein by reference for all purposes, are also known. These scanning systems include an optical train which directs a monochromatic or polychromatic light source to about a 5 micron (μm) diameter spot at its focal plane. In some cases, a photon counter detects the emission from the device in response to the light. The data collected by the photon counter represents one pixel or data point of the image. Thereafter, the light scans another pixel as a translation stage moves the device to a subsequent position.
As disclosed, these scanning confocal microscope systems provide high resolution by using an appropriate objective lens and large field of view by using appropriate translation stages. These translation stage-based confocal microscopes, however, obtain high resolution and field of view by sacrificing system throughput. As an example, an array of sequenced material using the pioneering fabrication techniques, such as those disclosed in U.S. Patent No. 5,143,854 (Pirrung et al.) and U.S. Patent Application Serial No. 08/143,312, incorporated herein by reference for all purposes, may have a density of about 105 sequences. Assuming that 36 pixels are required for each sequence, the image can take over at least 10 minutes to acquire.
From the above, it is apparent that improved methods and systems for imaging a sample are desired.
SUMMARY OF THE INVENTION
The present invention provides systems, methods and apparatus for detecting marked regions on substrate surfaces. In particular, the present invention provides methods and apparatus for scanning a substrate to obtain an image with high sensitivity and resolution at a high speed. The confocal scanning microscopes of the present invention combine the high scan rate of galvanometer based scanning microscopes with a sufficiently high resolution, sensitivity and a large enough field of view for imaging high density arrays of materials, such as those found in the fields of combinatorial chemistry and genetic analysis.
In one aspect, the present invention provides a system for detecting marked regions on a surface of a substrate, which comprises an excitation radiation source, and focusing optics for focusing the excitation radiation to regions on the surface of the substrate. A radiation direction system is also included for linearly scanning the focused excitation radiation across the surface of the substrate. A detector is positioned for detecting an emission from the substrate surface in response to the excitation radiation, and a data acquisition system records the amount of detected emission as a function of a position on the surface of the substrate from which the emission was detected.
In one embodiment, the focusing optics comprises an objective lens having a ratio of scanning field diameter to focused spot diameter of greater than 2000, preferably greater than 3000 and more preferably greater than 4000. Thus, the microscope is, for example, capable of focusing a laser beam to a spot having a diameter of about 3 microns at any point within a flat field having a length of about 14 mm. In addition, the objective lens has at least a 0.2 numerical aperture, preferably a 0.25 numerical aperture, which provides sufficient sensitivity to detect fluorescently marked regions on the substrate.
The radiation direction system preferably comprises a galvanometer mirror that scans the excitation radiation across the surface of the substrate. The objective lens has an external entrance pupil preferably located at or near the galvanometer mirror's pivot location. The mirror usually oscillates at a frequency of at least 7.5 Hz, preferably at least 20 Hz and more preferably at least 30 Hz. In this manner, the laser spot can usually be scanned across the substrate at velocities of about 5 image lines/second, preferably at least 10 images lines/second, and more preferably at least about 30 image lines/second. This allows the microscope to rapidly scan high density substrates, such as the polymer array substrates disclosed by Pirrung. It should be noted that the mirror may scan unidirectionally
(e.g., with a sawtooth wave) or bidirectionally (e.g., with a symmetric triangle wave) . In the latter case, the galvanometer frequency would generally be about half of the data aquisition speed in image lines/second. Accordingly, the frequency of the galvanometer in the latter case may be lower than 7.5 Hz in order to scan 5 image lines/second.
In another embodiment, the present invention also provides a system for detecting fluorescent regions on a surface of a substrate, which comprises an excitation radiation source, and first focusing optics for focusing the excitation radiation on the surface of the substrate in a focused spot having a diameter no greater than 10 μm, preferably less than 5 μm and more preferably about 3 μm. An oscillating or reciprocating radiation direction system scans the spot linearly across the surface of the substrate, with a focused travel distance of at least 10 mm and preferably about 14 mm. In one embodiment, an optical train separates fluorescence emitted from the surface of the substrate from the excitation radiation reflected from the surface. An autofocus system may also be included for automatically placing the surface of the substrate in a focal plane of the focusing optics. The present invention also provides methods of scanning substrates using the above systems. For example, in one aspect, the invention provides a method of scanning a polymer array having a plurality of different polymer sequences, each of the different polymer sequences being immobilized on a surface of a substrate in a different known location, to identify which polymer sequence on the array is bound by a fluorescent target molecule. The method comprises focusing an excitation radiation source upon the surface of the substrate, and scanning the excitation radiation across the surface of the substrate at a speed of at least 5 image lines/second. Fluorescent emissions are collected from the surface of the substrate in response to the excitation radiation. These fluorescent emissions are recorded as a function of a position on the surface of the substrate. The position on the surface indicates the polymer sequence on the array that is bound by the fluorescent target molecule.
A further understanding of the nature and advantages of the inventions herein may be realized by reference to the remaining portions of the specification and the attached drawings.
BRIEF DESCRIPTION OF THE DRAWINGS
Fig. 1 is a schematic illustration of one embodiment of a scanning system according to the present invention.
Fig. 2 illustrates one embodiment of a scanning system that includes a flow cell on which a substrate is mounted.
Fig. 3 is a schematic illustration of a computer based system for controlling and recording data from a scanning system according to the present invention. Fig. 4 is a schematic illustration of an alternate embodiment of a scanning system according to the present invention.
Fig. 5 is an enlarged view of a scanned image generated using a scanning system of the present invention.
DESCRIPTION OF THE PREFERRED EMBODIMENT
CONTENTS
I. Definitions
II. General a. Introduction b. Overview of the Imaging System
III. Detailed Description of One Embodiment of the Imaging System a. Detection Device b. Data Acquisition
IV. Detailed Description of a Second Embodiment of the Imaging System
I. Definitions
The following terms are intended to have the following general meanings as they are used herein:
1. Complementary: Refers to the topological compatibility or matching together of interacting surfaces of a probe molecule and its target. Thus, the target and its probe can be described as complementary, and furthermore, the contact surface characteristics are complementary to each other.
2. Probe: A probe is a surface-immobilized molecule that is recognized by a particular target. Examples of probes that can be investigated by this invention include, but are not restricted to, agonists and antagonists for cell membrane receptors, toxins and venoms, viral epitopes, hormones (e.g. , opioid peptides, steroids, etc.), hormone receptors, peptides, enzymes, enzyme substrates, cofactors, drugs, lectins, sugars, oligonucleotides, nucleic acids, oligosaccharides, proteins, and monoclonal antibodies.
3. Target: A molecule that has an affinity for a given probe. Targets may be naturally-occurring or manmade molecules. Also, they can be employed in their unaltered state or as aggregates with other species. Targets may be attached, covalently or noncovalently, to a binding member, either directly or via a specific binding substance. Examples of targets which can be employed by this invention include, but are not restricted to, antibodies, cell membrane receptors, monoclonal antibodies and antisera reactive with specific antigenic determinants (such as on viruses, cells or other materials), drugs, polynucleotides, nucleic acids, peptides, cofactors, lectins, sugars, polysaccharides, cells, cellular membranes, and organelles. Targets are sometimes referred to in the art as anti-probes. As the term targets is used herein, no difference in meaning is intended. A "Probe Target Pair" is formed when two macromolecules have combined through molecular recognition to form a complex.
II. General
A. Introduction
The present invention provides methods and apparatus for scanning a substrate to obtain a highly sensitive and resolved image at a high speed. The invention will have a wide range of uses, particularly, those requiring quantitative study of a microscopic region from within a larger region, such as 2 or 3 μm2 over 100 mm2. For example, the invention may find application in the field of histology (for studying histochemical stained and immunological fluorescent stained images) , or fluorescence in situ hybridization. In one application, the invention herein is used to image an array of probe sequences fabricated on a support.
High resolution scanning systems and methods, whether microscopic or macroscopic, have routinely been used in the electronics industries, e.g., in the semiconductor and microfabrication industries, to scan microfabricated electronics, e.g., microprocessors, microcircuitry, and the like. However, such scanning also has great utility in the field of combinatorial chemistry and genetic analysis. Specifically, high resolution scanning methods and devices can be used in the application of polymer arrays. These polymer arrays are generally made up of a large number of different polymer sequences that are coupled to the surface of a typically planar substrate. The substrate on which the polymer sequences are formed may be composed from a wide range of material, either biological, nonbiological, organic, inorganic, or a combination of any of these, existing as particles, strands, precipitates, gels, sheets, tubing, spheres, containers, capillaries, pads, slices, films, plates, slides, etc. The substrate may have any convenient shape, such as a disc, square, sphere, circle, etc. The substrate is preferably flat but may take on a variety of alternative surface configurations. For example, the substrate may contain raised or depressed regions on which a sample is located. The substrate and its surface preferably form a rigid support on which the sample can be formed. The substrate and its surface are also chosen to provide appropriate light-absorbing charac¬ teristics. For instance, the substrate may be a polymerized Langmuir Blodgett film, functionalized glass, Si, Ge, GaAs, GaP, Si02, SiN4, modified silicon, or any one of a wide variety of gels or polymers such as (poly)tetrafluoroethylene, (poly)vinylidenedifluoride, polystyrene, polycarbonate, or combinations thereof. Other substrate materials will be readily apparent to those of skill in the art upon review of this disclosure. In a preferred embodiment, the substrate is flat glass or silica. According to some embodiments, the surface of the substrate is etched using well known techniques to provide for desired surface features. For example, by way of the formation of trenches, v-grooves, mesa structures, or the like, the synthesis regions may be more closely placed within the focus point of impinging light. The surface may also be provided with reflective "mirror" structures for maximization of emission collected therefrom.
Surfaces on the solid substrate will usually, though not always, be composed of the same material as the substrate. Thus, the surface may be composed of any of a wide variety of materials, for example, polymers, plastics, resins, polysaccharides, silica or silica-based materials, carbon, metals, inorganic glasses, membranes, or any of the above- listed substrate materials. In one embodiment, the surface will be optically transparent and will have surface Si-OH functionalities, such as those found on silica surfaces.
These arrays are typically prepared in a manner that allows groups of each different polymer sequence to be coupled in a different known location on the substrate surface. The pioneering methods for synthesizing these arrays have been previously described. For example, methods of synthesizing arrays of polymers using light directed synthesis methods are described in U.S. Patent No. 5,143,854 to Pirrung et al., and U.S. Patent No. 5,405,783 to Fodor et al. , the complete disclosures of which are incorporated herein by reference. Additionally, mechanical methods for synthesizing these arrays are described in, e.g., U.S. Patent No. 5,384,261 to Winkler et al., the complete disclosure of which is incorporated herein by reference. Using these pioneering methods, the combination of photolithograpic and fabrication techniques may, for example, enable each probe sequence ("feature") to occupy a very small area ("site") on the support. In some embodiments, this feature site may be as small as a few microns or even a single molecule. For example, about 105 to 106 features may be fabricated in an area of only 12.8 mm2. Such probe arrays may be of the type known as Very Large Scale Immobilized Polymer Synthesis (VLSIPS™) . For ease of discussion, the arrays for use with the present invention are described in terms of arrays of different oligonucleotide sequences. However, it should be readily understood that a variety of different polymer array types are equally applicable to the devices of the invention. Furthermore, the scanners of the present invention may also be used in the reflectance mode to scan non-fluorescent surfaces.
In those applications using polymer arrays, such arrays may be prepared having all nucleotide sequences of a given length, composed of the basis set of monomers. For example, an array of oligonucleotides containing all possible sequences of length n which is made up of the basis set of four nucleotides contains up to 4n oligonucleotides on its surface. For an array of 8mer or lOmer oligonucleotides, such arrays may include upwards of about 65,536 and 1,048,576 different oligonucleotides, respectively. Generally, where it is desired to produce arrays having all possible polymers of length n, a simple binary masking strategy can be used, as described in U.S. Patent No. 5,143,854 to Pirrung. Alternate masking strategies can produce arrays of probes which contain a subset of polymer sequences, i.e., polymers having a given subsequence of monomers, but are systematically substituted at each position with each member of the basis set of monomers. In the context of oligonucleotide probes, these alternate synthesis strategies may be used to lay down or "tile" a range of probes that are complementary to, and span the length of a given known nucleic acid segment. The tiling strategy will also include substitution of one or more individual positions within the sequence of each of the probe groups with each member of the basis set of nucleotides. These positions are termed "interrogation positions." By reading the hybridization pattern of the target nucleic acid, one can determine if and where any mutations lie in the sequence, and also determine what the specific mutation is by identifying which base is contained within the interrogation position. Tiling methods and strategies are discussed in substantial detail in Published PCT Application No. 95/11995, the complete disclosure of which is incorporated herein by reference in its entirety for all purposes.
Tiled arrays may be used for a variety of applications, such as identifying mutations within a known oligonucleotide sequence or "target". Specifically, the probes on the array will have a sub-sequence which is complementary to a known nucleic acid sequence, but wherein at least one position in that sequence has been systematically substituted with the other three nucleotides. See, e.g., U.S. Patent No. 5,527,681, the complete disclosure of which is incorporated herein by reference.
Generally, the sample nucleic acid for which sequence information is desired is contacted with the array. This "target" sequence is typically labeled with a detectable group such as a fluorescent moiety, i.e., fluorescein or rhodamine. Following hybridization of the target to the array, one can detect the position on the array to which the target sequence binds by scanning the surface of the array for fluorescence. The surface is typically scanned by directing excitation radiation at the surface to activate the fluorescent labeling group in the target, which in turn emits a fluorescent response radiation. The fluorescent response radiation is detected and assigned to the region from which it originated. By knowing the position from which the fluorescence originates, one can identify the sequence to which the target binds.
Although generally used herein to define separate regions containing differing polymer sequences, the term "feature" generally refers to any element, e.g. , region, structure or the like, on the surface of a substrate. Typically, substrates to be scanned using the scanning systems described herein, will have small feature sizes, and consequently, high feature densities on substrate surfaces.
For example, individual features will typically have at least one of a length or width dimension that is no greater than 100 μm, and preferably, no greater than 50 μm, and more preferably greater than about 20 μm. Thus, for embodiments employing substrates having a plurality of polymer sequences on their surfaces, each different polymer sequence will typically be substantially contained within a single feature.
The probe arrays will have a wide range of applications. For example, the probe arrays may be designed specifically to detect genetic diseases, either from acquired or inherited mutations in an individual DNA. These include genetic diseases such as cystic fibrosis, diabetes, and muscular dystrophy, as well as acquired diseases such as cancer (P53 gene relevant to some cancers) , as disclosed in U.S. Patent Application Serial Number 08/143,312, already incorporated by reference.
Genetic mutations may be detected by a method known as sequencing by hybridization. In sequencing by hybridization, a solution containing one or more targets to be sequenced (i.e., samples from patients) contacts the probe array. The targets will bind or hybridize with complementary probe sequences. Generally, the targets are labeled with a fluorescent marker, radioactive isotopes, enzymes, or other types of markers. Accordingly, locations at which targets hybridize with complimentary probes can be identified by locating the markers. Based on the locations where hybridization occur, information regarding the target sequences can be extracted. The existence of a mutation may be determined by comparing the target sequence with the wild type.
The interaction between targets and probes can be characterized in terms of kinetics and thermodynamics. As such, it may be necessary to interrogate the array while in contact with a solution of labeled targets. Consequently, the detection system must be extremely selective, with the capacity to discriminate between surface-bound and solution- born targets. Also, in order to perform a quantitative analysis, the high-density volume of the probe sequences requires the system to have the capacity to distinguish between each feature site.
B. Overview of the Imaging System
The devices of the present invention generally employ a scanning device which rapidly sweeps an activation radiation beam or spot across the surface of the substrate. The devices also include focusing optics for focusing the excitation radiation onto the surface of the substrate in a sufficiently small area to provide high resolution of features on the substrate, while simultaneously providing a wide scanning field. An image is obtained by detecting the electromagnetic radiation emitted by the labels on the sample when the labels are illuminated. In one embodiment, fluorescent emissions are gathered by the focusing optics and detected to generate an image of the fluorescence on the substrate surface. In preferred aspects, the devices of the invention further employ confocal detection systems to reduce or eliminate unwanted signals from structures above and below the plane of focus of the excitation radiation, as well as autofocus systems to focus both the activation radiation on the substrate surface and the emitted radiation from the surface. Generally, the excitation radiation and response emission have different wavelengths. Filters having high transmission in the label's emission band and low transmission in the excitation wavelength may be utilized to inhibit the detection of undesirable emission. These generally include emission from out-of-focus planes or scattered excitation illumination as potential sources of background noise.
In operation, the devices of the present invention include one or more sources of excitation radiation. Typically, these source(s) are immobilized or stationary point light sources, e.g., lasers such as argon, helium-neon, diode, dye, titanium sapphire, frequency-doubled diode pumped Nd:YAG and krypton. Typically, the excitation source illuminates the sample with an excitation wavelength that is within the visible spectrum, but other wavelengths (i.e., near ultraviolet or near infrared spectrum) may be used depending on the application (i.e., type of markers and/or sample). In some embodiments, the sample is excited with electromagnetic radiation having a wavelength at or near the absorption maximum of the species of label used. Exciting the label at such a wavelength produces the maximum number of photons emitted. For example, if fluorescein (absorption maximum of 488 nm) is used as a label, an excitation radiation having a wavelength of about 488 nm would induce the strongest emission from the labels.
In instances where a multi-labeling scheme is utilized, a wavelength which approximates the mean of the various candidate labels' absorption maxima may be used. Alternatively, multiple excitations may be performed, each using a wavelength corresponding to the absorption maximum of a specific label. Table I lists examples of various types of fluorophores and their corresponding absorption maxima.
Figure imgf000018_0001
The excitation radiation from the point source is directed at a movable radiation direction system which rapidly scans the excitation radiation beam back and forth across the surface of the substrate. A variety of devices may be employed to generate the sweeping motion of the excitation radiation. For example, resonant scanner or rotating polygons, may be employed to direct the excitation radiation in this sweeping fashion. Generally, however, galvanometer devices are preferred as the scanning system. As used herein, the term "galvanometer" refers to devices that employ a servo motor to oscillate or rotate a mirror over a restricted, predefined range, which is typically less than 90°. This generates a rapidly sweeping or rastering beam reflecting from the galvanometer mirror, which is then directed at and swept across the surface of a substrate that is to be scanned. Typically, an optical train may be employed between the activation source and the galvanometer mirror to assist in directing, focusing or filtering the radiation directed at and reflected from the galvanometer mirror.
The galvanometers employed in the devices and systems of the present invention typically sweep a scanning spot across the substrate surface at an oscillating frequency that is typically greater than about 7.5 Hz, preferably greater than about 20 Hz and more preferably, greater than about 30 Hz. With this frequency, the spot can typically be scanned across the substrate at a velocity of at least about 20 lines/second, preferably in about 5 image lines/second, preferably at least 10 images lines/second, and more preferably at least about 30 image lines/second. It should be noted that the mirror may scan unidirectionally (e.g. , with a sawtooth wave) or bidirectionally (e.g., with a symmetric triangle wave) . In the latter case, the galvanometer frequency would generally be about half of the data aquisition speed in image lines/second. Accordingly, the frequency of the galvanometer in the latter case may be lower than 7.5 Hz in order to scan 5 image lines/second. The activation radiation is then directed through focusing optics to focus the sweeping beam at the surface of the array which is used to interrogate the target sequence. These same focusing optics also collect emitted fluorescence from the substrate surface for subsequent detection. The objective lens is preferably selected to provide high resolution, as determined by the focused spot size, while still allowing a wide scanning field. "Focused spot size" is defined by the diameter of the focused activation spot on the surface of the substrate at 1/e2 intensity points. "Scanning field" is defined as the length of travel of the focused activation beam or "spot," in one dimension parallel to the direction of travel of the sweeping beam. The confocal system of the present invention generally provides a focused spot size having a diameter no greater than about 10 μm, preferably no greater than about 5 μm, and more preferably about 3 μm. In addition, these systems have a scanning field with a dimension parallel to the direction of the sweep of the activation beam that is usually greater than about 10 mm and more preferably about 14 mm. For ease of discussion, the combined measurement of resolution and effective scanning field are provided as a ratio of the size of the effective field of view of the objective to the size of the focused spot from the scanner. For example, a scanner having a scanning field of 2 mm and a focused spot size of 10 μm would have a ratio of 200 (2mm/0.010 mm). While a scanner having an effective scanning field of 14 mm with a focused spot size of 3 μm would have a ratio of 4666. In general, the scanning system of the present invention usually has a ratio of size of effective field of view to size of focused spot of at least about 2000, preferably greater than about 3000, and more preferably greater than about 4000. It should be noted that "scanning field" generally refers to the direction in which the galvanometer mirror scans the laser spot across the substrate. The translation stage moves the substrate in the direction orthogonal to the scanned direction, and this direction may be larger than 14 mm. In one embodiment of the present invention, high fluorescence collection efficiency is achieved by providing an objective lens having a numerical aperture that is at least about 0.2. In particularly preferred aspects, the numerical aperture is at least about 0.25. This translates to an F/number of about F/2.5 to F/2.
In addition to providing smaller spot size while not sacrificing scanning field size, the focusing optics of the scanning system may include a telecentric objective lens. This allows the activation beam to strike the scanning surface at an angle that is substantially normal to that surface, across the entire scanning surface, i.e., regardless of where the input beam is propagating through the lens relative to the lens axis. By "substantially normal" is meant an angle of incidence of approximately 0°, e.g., between 0° and 5°, and preferably from 0° to 2°.
As the activation radiation spot is swept across the surface of the substrate, it activates any fluorescent groups that remain upon the surface, e.g., those that are bound to the surface. The activated groups emit a response radiation or emission which is then collected by the objective lens and directed back through the optical train via the servo mounted mirror. In order to avoid the detrimental effects of reflected excitation radiation upon the detection of the fluorescence, dichroic mirrors or beam splitters may be included in the optical train. These dichroic beam splitters or mirrors are reflective to radiation in the wavelength of the excitation radiation while transmissive to radiation in the wavelength of the response radiation. For example, where an Argon laser is used as the point energy source, it will typically generate activation radiation having a wavelength of about 488 nm. Fluorescence emitted from an activated fluorescein moiety on the other hand will typically have a wavelength between about 515 and 545 nm. As such, dichroic mirrors may be included which transmit light having a wavelength greater than 515 nm while reflecting light of shorter wavelengths. This effectively separates the excitation radiation reflected from the surface of the substrate from the response radiation emitted from the surface of the substrate. Similarly, additional dichroic mirrors may be used to separate signals from label groups having different response radiation wavelengths, thereby allowing simultaneous detection of multiple fluorescent indicators, and thus, simultaneous interrogation of a single array with multiple target sequences. Specifically, a first target sequence may be labeled with a shorter wavelength fluorescent label, e.g., fluorescein, while a second target sequence is labeled with a longer wavelength fluorescent label, e.g., carbocyanin dyes such as CY3 which emit response radiation in the range of 550 to 600 nm. The response radiation from each of these targets may be separated and individually detected through the inclusion of additional dichroic beam splitters and detectors. Following separation of the response radiation from the reflected excitation radiation, the response radiation or fluorescence is then directed at a detector, e.g., a photomultiplier tube, to measure the level of response radiation and record that level as a function of the position on the substrate from which that radiation originated. Typically, the response radiation is focused upon the detector through a spatial filter such as a confocal pinhole. Such spatial filters reduce or eliminate unwanted signals from structures above and below the plane of focus of the excitation radiation. Additionally, the device may incorporate a bandpass filter between the dichroic mirror and the detector to further restrict the wavelength of radiation that is delivered to the detector. As noted above, the reflected excitation radiation may generally be used in an autofocusing system in the devices described herein. In particular, the reflected excitation radiation may also be directed at a detector, e.g., a photodiode, that is preferably located behind another spatial filter, i.e., a confocal pinhole.
The substrate surface may be moved closer to and further from the objective lens using, e.g., a translation stage upon which the substrate is mounted. As the substrate is moved out of focus, the amount of reflected excitation radiation contacting the photodiode will be reduced. As the substrate is brought back into focus, this amount will increase, reaching a maximal value when the substrate is in focus. Control for the autofocus system is generally supplied by an appropriately programmed computer, which moves the translation stage in response to the input from the photodiode, until a maximum is reached. Generally, this computer is the same computer which receives, compiles and stores the input from the fluorescence detector or photomultiplier tube, to produce an output of fluorescence as a function of position on the array, typically in the form of a numerical representation or a scan image. Examples of autofocus confocal systems for use in fluorescent scanning devices are generally described in commonly assigned, co- pending U.S. Patent Application Serial No. 08/195,889, filed February 10, 1994, the complete disclosure of which is incorporated herein by reference for all purposes. These autofocus systems generally permit the focusing of light reflected from a weakly reflecting surface, e.g., the wet side of a glass substrate, even in the vicinity of a strongly reflecting surface, e.g., the dry side of a glass substrate, as well as being capable of focusing on a featureless surface, e.g., smooth glass. III. Detailed Description of One Embodiment of the Imaging System. A. Detection Device
Fig. 1 is an optical block diagram illustrating the imaging system 100 according to the present invention.
Typically, imaging system 100 is used to obtain images of oligonucleotide probe arrays to which fluorescently labelled DNA or RNA is bound. It might also be used for other applications, such as wafer or mask inspection, or for imaging of polypeptide or other polymer arrays, electrophoresis gels, or biological specimens. As shown in Fig. 1, a beam of excitation radiation, e.g., 488-nm light, from a laser 102 is partially reflected and partially transmitted by a beamsplitter 104. The reflected portion of the beam impinges upon a photodetector 131 (optional) , which is typically a photodiode used as a laser power monitor. The portion of the beam that is transmitted through beamsplitter 104 is reflected by dichroic beamsplitters 106 and 108 and transmitted through lenses 111 and 112, which provide a telescope to expand the beam emitted by laser 102 and to demagnify the collimated fluorescence obtained from the surface of the substrate. In an exemplary embodiment, lens ill has a focal length of 20 mm and lens 112 has a focal length of 80 mm. Other focal lengths and focal length ratios can be used, but system performance (confocality, resolution, etc.) may be affected. The expanded laser beam is reflected by mirror 114 and focused by lens 116, which is described in more detail below.
Mirror 114, which typically is an 18 mm x 28 mm octagon or ellipse, is a galvanometer mirror that oscillates (i.e., scans) at a rate of typically several Hertz or several tens of Hertz. In preferred aspects, the galvanometer mirror oscillates at frequencies greater than 7.5 Hz, preferably about 20 Hz and more preferably about 30 Hz. A resonant scanner or rotating polygonal mirror could be used instead of a galvanometer. Typically, imaging system 100 obtains an image of a 2-dimensional area by scanning the laser beam in one dimension while moving the sample in an orthogonal direction. Optimally, sample 118 is positioned such that the plane of interest (e.g. the surface to which labelled target molecules are bound) is located in the plane where the laser light is focused. Light remitted by sample 118 is collected by lens 116, reflected by mirror 114, and transmitted through lenses 112 and 111. Remitted light may consist of fluorescence, phosphorescence, specular reflection, diffuse reflection, Raman scattering, etc. Remitted light having a wavelength greater than 555 nm passes through dichroic beamsplitter 108 and is focused by lens 134 onto pinhole 136. Remitted light having a wavelength between 515 and 555 nm is reflected by beamsplitter 108, passes through dichroic beamsplitter 106, and is focused by a lens 120 onto a pinhole 122. The light that is transmitted through pinholes 136 and 122 impinges upon filters 138 and 124, respectively; the light that passes through these filters impinges upon photodetectors 140 and 126, respectively. Filter 138 transmits light having wavelengths between 555 and 607 nm, and filter 124 transmits light having wavelengths between 515 and 545 nm. Remitted light having wavelengths less that 515 nm is reflected by beamsplitters 108 and 106 and partially reflected by beamsplitter 104; the light reflected by beamsplitter 104 is focused by a lens 132 onto a pinhole 128. The light that is transmitted through pinhole 128 impinges upon a photodetector 130.
Pinholes 136, 122, and 128 are typically confocal pinholes. Specifically, remitted light that originates from the location on sample 118 where the laser beam is focused is maximally transmitted through the pinholes, while light that originates from other locations is not. Lenses 134, 120, and 132 preferably have focal lengths of 50 mm. Pinholes 136 and 122 preferably have diameters of 100 microns; pinhole 128 preferably has a diameter of 50 microns. Other lens focal lengths and pinhole diameters can be used, but system performance (confocality, sensitivity to misalignment, etc.) may be affected.
If lens 116 is not fully corrected for lateral color, the remitted light that is focused onto pinholes 136 and 122 might move laterally (across the plane of the pinhole) as mirror 114 is scanned. In this case it might be advantageous to use pinholes that are elliptical or rectangular rather than round, or to use slits instead of pinholes. For example, lens 116 in the present scanner has about 3 microns of lateral color at the edges of the scan field; consequently, the remitted light can move laterally by about 40 microns across the pinholes. Reasonable sizes for pinholes 136 and 122 in this case might be approximately 75 microns by 125 microns.
Photodetectors 140 and 126 are typically photomultipliers and are typically intended for the detection of relatively weak signals such as fluorescence. Various light detectors may be used, including photodiodes, avalanche photodiodes, phototransistors, vacuum photodiodes, photomultiplier tubes, and other light detectors.
Typically, photodetector 130 is used to aid in focusing the sample prior to scanning or while it is being scanned. The amount of reflected laser light reaching photodetector 130 is a maximum when the laser beam is focused onto the surface of sample 118. Focusing can be done either manually, or by a motorized translation stage under computer control. Frequently, sample 118 is a glass slide mounted in a liquid-filled flow cell (see Fig. 2) and the surface of interest is the second surface of the glass, i.e. the glass- liquid interface. Typically the reflection from this surface is much weaker than the reflection from the first surface of the glass, i.e. the glass-air interface, but the system is sufficiently confocal that the reflection peaks from the 2 surfaces are well separated.
The system described above is particularly useful in the detection of fluorescence from targets that are labelled with dyes such as fluorescein and phycoerythrin. Other dyes (rhodamines, carbocyanines, etc.) can be used, but laser 102, beamsplitters 108 and 106, and filters 138 and 124 may need to be altered depending on the dyes' absorption and emission spectra. In some cases, target molecules may be labeled with particles or very large molecules that scatter laser light but do not fluoresce. In these cases, it may be desirable to detect diffusely scattered light but not specularly reflected laser light. An annular aperture can be placed between beamsplitter 104 and lens 132. If the inner diameter of the annulus is selected properly, this aperture will block specularly reflected laser light while transmitting diffusely scattered laser light, which can then be detected by photodetector 130.
Objective lens 116 was designed to meet a number of specifications, and may be obtained from, e.g. , Special Optics, Part No. 55-S30-15(Wharton, NJ) . The objective lens is usable with a galvanometer (or resonant scanner, rotating polygon, etc.), i.e. with an entrance pupil that is external to the lens and is located at or near the galvanometer mirror's pivot location. The lens is also capable of focusing a TEM00-mode laser beam having a wavelength of 488 nm to a spot having a diameter no greater than 3 microns at the 1/e2 intensity points, and should be capable of doing this at any point within a flat field having a diameter no less than 14 mm.
Objective lens 116 is within 2 degrees of telecentric, i.e. the transmitted laser beam should be parallel to the optical axis of the lens to within 2 degrees. (If the lens is not telecentric or nearly so, specularly reflected light from near the edges of the field might miss the lens and therefore not be detected.) In addition, lens 116 was designed so that the F/number would be no greater than f/2, i.e. numerical aperture no less than 0.25, with no vignetting even at the edges of the field. (F/number is a measure of the lens's light-gathering ability and is important if the lens is to be used for the detection of weak signals) . Objective lens 116 is capable of focusing a polychromatic beam, having wavelengths between 488 and 600 nm and filling the entrance pupil, to a spot having a diameter no greater than 10 microns, and is capable of doing this at any point within a flat field having a diameter no less than 14 mm. Objective lens 116 preferably has a focal length of about 30 mm. A lens having a different focal length, but still meeting the above specifications could be readily designed and would be acceptable for the purposes described herein. Lens 116 can focus a 488 nm laser beam to a diffraction-limited spot when the ratio of input beam diameter to lens focal length is less than approximately 0.21, e.g. it can focus a 6.2 mm diameter beam to a 3 micron diameter spot. If a larger focal spot is desired, the input beam diameter can be made smaller, e.g. by changing the ratio of the focal lengths of lenses 111 and 112 or by demagnifying the beam between laser 102 and beamsplitter 104.
Lens 116 was designed to be able to image a 14 mm field with 3 micron resolution and 0.25 numerical aperture; however, the design can easily be scaled up, e.g. to image a 28 mm field with 6 micron resolution and 0.25 numerical aperture (or potentially, to image a 280 mm field with 60 micron resolution and 0.25 numerical aperture, although the lens would be extremely large and costly) . This scale-up involves merely multiplying the thicknesses, diameters, and radii of curvature of all of the elements of lens 116 by a constant. Similarly, the entire scanner can be scaled up for imaging of larger areas by multiplying all of the lens diameters and focal lengths, pinhole diameters, and mirror diameters by the same factor.
B. Data Acquisition
As shown in Fig. 3, imaging system 300 includes a galvanometer 306 (to which a mirror 114 is attached) and a galvanometer driver board 305, which can be obtained from General Scanning Inc. (Watertown, MA) . Galvanometer 306 is a model M2T. Other suitable galvanometers and driver boards are available, e.g. from Cambridge Technology Inc. (Watertown, MA) . The input to driver board 305 is a voltage waveform from arbitrary waveform generator 304 (Keithley Metrabyte model PCIP-AWFG, Taunton, MA) , which is installed in an ISA slot in computer 302. Circuitry on board 305 attempts at every instant to force galvanometer 306 to whatever angular position is commanded by waveform generator 304 (the desired angular position is linearly related to the waveform voltage) . Waveform generator 304, after being programmed, can generate a waveform indefinitely without further intervention by computer 302. The waveform used is typically a sawtooth wave: e.g. if the waveform period is 33.3 msec, the voltage ramps up linearly for 25 msec, during which time data are acquired as the laser beam sweeps across the field; during the next 8.3 msec the voltage returns to its initial value and the laser beam retraces. Other waveforms such as sine waves or symmetric triangle waves could be used. Various methods of waveform generation that do not require the PCIP-AWFG waveform generator are known. Currents from photomultipliers 320 and 321 and from photodiode 322 are converted to voltages by either transimpedance amplifiers or load resistors, optionally followed by voltage amplifiers. Simple op-amp circuits for this purpose are well known. The voltages are then low-pass filtered by filters 310, 311, and 312, e.g., programmable 4- pole Bessel filters (Frequency Devices model 824L8L-6, Haverhill, MA) . Filter cutoff frequencies are digitally programmable in 400-Hz steps from 400 Hz to 102.4kHz and are set by digital input-output board 314 (Computer Boards Inc. model CIO-DI024, Mansfield, MA) installed in computer 302. Similar digital I/O boards are available from several other manufacturers. Filter cutoff frequencies are set by software so that each filter's output rise time in response to a step input is approximately equal to the time between A/D conversions. For example, if A/D conversions are 6 microseconds apart, the cutoff frequencies are set to 66 kHz. A variety of other types of filters (filters with more or fewer poles of different transfer functions (e.g. Butterworth) , fixed-frequency filters, or simple RC filters) could be used instead.
Filter outputs are digitized by a 12-bit, 330-kHz analog-to digital converter on data acquisition board 308 (Computer Boards Inc. model CIO-DAS16/330) . Similar data acquisition boards are available from several other manufacturers. Digitized data are displayed by computer 302, typically in the form of a gray-scale image, and are written to the computer's hard disk for subsequent data analysis. If the internal clock on data acquisition board 308 is used to trigger A/D conversions, the images obtained may be slightly distorted. A/D conversions that are equally far apart in time are not equally far apart in space, i.e., the velocity of the laser beam across sample 118 is not exactly constant, partly because lens 116 is not an ideal f-theta lens, and partly because of non-ideal behavior by driver board 305 and galvanometer 306. Therefore, A/D conversions are preferably triggered by waveform generator 304, which has digital output channels in addition to its analog output channel. When waveform generator 304 is appropriately programmed and its digital output pulses are appropriately spaced, image distortion can be eliminated.
Typically 4096 A/D conversions are performed per scan line, and if the user wants fewer than 4096 pixels/line the A/D conversions are binned by software in groups of 2 or more. For example, if the length of a scan line is 14 mm, the laser beam moves 3.4 microns between one A/D conversion and the next; and if, for example, the user chooses a pixel size of 13.6 microns, software adds the numbers from 4 A/D conversions for each pixel. One version of the software allows 8192 A/D conversions per scan line. The utility of doing more A/D conversions per line diminishes as the pixel size becomes less than the laser spot size.
Translation stages 332 (typically a 2-axis or 3-axis set of stages) are controlled by indexer 330. Two axes of translation are usually used: a scan axis (parallel to the plane of the sample and perpendicular to the direction of motion of the laser spot) , and a focus axis (parallel to the optical axis of lens 116) . A third axis orthogonal to the other two may also be desirable. Computer 302 sends commands to indexer 330, and receives information about translation- stage status from indexer 330, e.g., using an RS-232 interface. An indexer that communicates with computer 302 using another interface, such as a GPIB or ISA interface, could be used instead. Translation stage speed is set such that the distance that sample 118 moves per scan line is equal to the desired pixel size. Software allows the user to control various scan parameters, including the size of the area to be scanned, the pixel size, the scan speed, and which output or outputs to digitize (either of the photomultiplier outputs, or the photodiode output, or a combination thereof) . Default parameters are 14 mm x 14 mm scan area, 10.2 micron pixel size, and scan speed of 30 lines/second. With these parameters, an image contains 1365 x 1365 pixels and can be acquired in about 45 seconds. These parameters are usually suitable for users who need to image 12.8 mm x 12.8 mm oligonucleotide arrays having features that are approximately 100 μm x 100 μm. Other scan parameters are suitable in other circumstances. The source code for this software is enclosed in Appendix A, of which is hereby incorporated herein by reference for all purposes. For example, if the features are 400 μm x 400 μm the user might choose a pixel size of 27.2 microns. In this case an image of a 14 mm x 14 mm area contains 512 x 512 pixels and can be obtained in about 17 seconds. On the other hand, if the features are 30 μm x 30 μm, the user might choose a pixel size of 3.4 microns. In this case an image of a 14 mm x 14 mm area contains 4096 x 4096 pixels and can be acquired in about 2 minutes and 16 seconds. If an image with adequate signal-to noise ratio is not obtained at a scan speed of 30 lines/second, the user might choose to reduce the scan speed, for example to 7.5 lines/second (quadrupling the scan time approximately quadruples the number of photons detected per pixel, and therefore in many cases approximately doubles the signal-to-noise ratio) .
Scanned areas can vary from the 14 mm x 14 mm dimensions described above, and need not be square. For example, software options can allow the user to reduce the length of a scan line, e.g., to as little as 4 mm or smaller if a user so desired, with a pixel size as small as 1 micron, and to set the number of lines per scan independently of the other scan parameters. In the galvanometer-scanned direction, a single scan can not cover a region that is larger than the field of view of lens 116, e.g., 14 mm. However, the system may cover a region that is several inches wide in the orthogonal direction (limited only by the length of travel of the translation stage) . Other software options allow users automatically to scan a chip several times, with any desired time interval between scans (useful for certain kinetics experiments) , and automatically to scan several different chips on a wafer (useful for wafer inspection prior to dicing and packaging) . Another option allows users to choose between 2 different output file formats: one format which saves "raw" data, with 2 bytes per pixel; and a space-saving format which saves "scaled" data, with one byte per pixel. A scaled data value is directly proportional to the square root of the raw data value. A lookup table for converting from scaled data back to raw data is stored in the file's header.
Referring to Fig. 2, a system and method for holding sample 118 within a flow cell will now be described. It should be clearly understood that the present invention is not limited to a flow cell. For example, the sample 118 may be part of a packaged chip, such as a diced chip glued into a disposable plastic package. A more complete description of a packaged chip can be found in commonly assigned, co-pending applications Serial Nos. 08/485,452, filed June 7, 1995 (Attorney Docket No. 16528X-006910) , the complete disclosures of which are incorporated herein by reference for all purposes. As shown, imaging system 100 further includes a body
422 for holding a support 430 containing the sample on a surface 431. In some embodiments, the support may be a microscope slide or any surface which is adequate to hold the sample. The body 422, depending on the application, may be a flow cell having a cavity 423. The flow cell, for example, may be employed to detect reactions between targets and probes. In some embodiments, the bottom of the cavity may comprise a light absorptive material so as to minimize the scattering of incident light.
In embodiments utilizing the flow cell, surface 431 is mated to body 422 and serves to seal cavity 423. The flow cell and the substrate may be mated for sealing with one or more gaskets. In one embodiment, the substrate is mated to the body by vacuum pressure generated by a pump 452. Optionally, the flow cell is provided with two concentric gaskets and the intervening space is held at a vacuum to ensure mating of the substrate to the gaskets. Alternatively, the substrate may be attached by using screws, clips, or other mounting techniques. When mated to the flow cell, the cavity encompasses the sample. The cavity includes an inlet port 421 and an outlet port 420. A fluid, which in some embodiments contains fluorescently labeled targets, is introduced into the cavity through inlet port 421. A pump 453, which may be a model no. B-120-S made by Eldex Laboratories, circulates fluids into the cavity via inlet 421 port and out through outlet port 420 for recirculation or disposal. Alternatively, a syringe, gas pressure, or other fluid transfer device may be used to flow fluids into and through the cavity.
Optionally, pump 453 may be replaced by an agitation system that agitates and circulates fluids through the cavity. Agitating the fluids shortens the incubation period between the probes and targets. This can be best explained in terms of kinetics. A thin layer, known as the depletion layer, is located above the probe sample. Since targets migrate to the surface and bind with the probe sequences, this layer is essentially devoid of targets. However, additional targets are inhibited from flowing into the depletion layer due to finite diffusion coefficients. As a result, incubation period is significantly increased. By using the agitation system to dissolve the depletion layer, additional targets are presented at the surface for binding. Ultrasonic radiation and/or heat, shaking the holder, magnetic beads, or other agitating technique may also be employed.
In some embodiments, the flow cell is provided with a temperature controller 450 for maintaining the flow cell at a desired temperature. Since probe/target interaction is sensitive to temperature, the ability to control it within the flow cell permits hybridization to be conducted under optimal temperature. The temperature controller 450 may include a circulating bath, a refrigerated air circulating device, resistance heater, peltier device (thermoelectric cooler) , or other temperature controller may be implemented.
As previously mentioned, the flow cell is oriented to maintain the substrate perpendicular to the optical axis of the collection optics. According to one embodiment, flow cell 422 may mounted to a translation stage 424 for moving the flow cell in an orthogonal direction relative to the optical path. The flow cell may be mated to the translation stage by vacuum pressure generated by pump 452. Alternatively, screws, clips or other mounting techniques may be employed to mate the flow cell to the translation stage.
IV. Detailed Description of an Alternative κm>»odiment of the Imaging System.
An optical block diagram of a second embodiment of imaging system 400 is shown in Fig. 4. As depicted, imaging system 400 comprises components which are common to the system described in Fig. 1. In this scanner, lens 116 is intended for use at a finite rather than an infinite conjugate ratio, and therefore lens 112 is omitted. This scanner uses only a single pinhole 401, and all of the remitted light passes through this pinhole 401 before being separated by the various dichroic beamsplitters. This scanner has 2 additional dichroic beamsplitters, spectral filters, and photomultipliers, and therefore can be used for 4-channel rather than merely 2-channel fluorescent scanning.
As discussed above in the previous embodiment, a beam of excitation radiation, e.g., 488-nm light, from a laser 102 is partially reflected and partially transmitted by a beamsplitter 104. The reflected portion of the beam impinges upon a photodetector 131 (optional) , which is typically a photodiode used as a laser power monitor. The portion of the beam that is transmitted through beamsplitter 104 is reflected by dichroic beamsplitter 106 and transmitted through lens 111, focused onto pinhole 401, and expanded to the desired diameter at the entrance pupil lens 116. The beam of excitation radiation is focused by lens
116 and scanned across the sample 118 by galvanometer mirror 114, as discussed above. Light remitted by sample 118 is collected by lens 116, reflected by mirror 114, and focused onto confocal pinhole 401. Light that is transmitted through pinhole 401 is collimated by lens 111. Remitted light having wavelengths less that 515 nm is reflected by beamsplitter 106 and partially reflected by beamsplitter 104; the light reflected by beamsplitter 104 impinges upon a photodetector 130. Remitted light having a wavelength above 515 passes through beamsplitter 106 and is sent to one of the four channels 402, 404, 406, 408 depending on its wavelength. For example, imaging system 300 may be constructed such that remitted light having a wavelength between 515 and 545 is reflected by dichroic beamsplitter 410, and passes through a filter 412 onto photodetector 414. Remitted light having a wavelength between 545 and 570 nm, for example, may be reflected by dichroic beamsplitter 416, where it passes through filter 418 and onto photodetector 420. Similarly, beamsplitter 422 is constructed to reflect remitted light having wavelengths between 570 and 595 nm through filter 423 onto photodetector 426. Remitted light having wavelengths greater than 595 nm are transmitted through beamsplitter 422, to pass through filter 425 onto photodetector 424.
The present invention is further illustrated by the following example. This example is merely to illustrate aspects of the present invention and is not intended as limitations of this invention.
EXAMPLE
A tiling array was designed for the entire human mitochondrial genome. See M. Chee et al., "Accessing genetic information with high-density DNA arrays", Science vol. 274, pgs. 610-614 (1996). The array contained 134,688 different probe sequences, each occupying a separate 35 μm feature. The overall array dimensions were 1.28 cm X 1.28 cm. Using long range PCR and RNA polymerase promoter tagged primers, a 16.3 kb fragment of mtDNA was amplified directly from genomic DNA samples. Labeled 16.3 kb RNA targets were prepared by in vitro transcription from the PCR amplicons and hybridized to the array.
A scanning system as described above was used to scan the array and a 4096X4096 pixel image was obtained, each pixel representing 3.4 μm. The entire array was scanned in under three minutes. A close-up view of a scanned image of this array is shown in Figure 5. The scanned image shown covers a 1.5 mm X 1.5 mm segment of 14 mm X 14 mm image of a 12.8 mm X 12.8 mm array.
While the foregoing invention has been described in some detail for purposes of clarity and understanding, it will be clear to one skilled in the art from a reading of this disclosure that various changes in form and detail can be made without departing from the true scope of the invention. All publications and patent documents cited in this application are incorporated by reference in their entirety for all purposes to the same extent as if each individual publication or patent document were so individually denoted.

Claims

WHAT IS CLAIMED IS;
1. A system for detecting marked regions on a surface of a substrate, the system comprising: an excitation radiation source; a focusing system for focusing radiation from said excitation radiation source onto a selected region of said surface of said substrate, said focusing system including an objective lens having a ratio of scanning field diameter to focused spot diameter of greater than about 2000, and a numerical aperture greater than about 0.2; a radiation direction system for scanning said focused excitation radiation across said surface of said substrate at a rate of at least 5 image lines/second; a detector for detecting an emission from said surface of said substrate in response to said excitation radiation, wherein said objective lens receives said emission and transmits the emission to the detector; and a data acquisition system for recording an amount of said emission detected as a function of a position on said surface of said substrate from which said emission was emitted.
2. The system of claim 1, wherein said focusing system has a ratio of scanning field diameter to focused spot diameter of greater than 3000.
3. The system of claim 1, wherein said focusing system has a ratio of scanning field diameter to focused spot diameter of greater than 4000.
4. The system of claim 1, wherein said focusing system focuses said excitation radiation in a spot having a diameter of less than about 10 μm.
5. The system of claim l, wherein said focusing system focuses said excitation radiation on said surface of said substrate in a spot having a diameter of less than about 5 μm.
6. The system of claim 1 wherein said focusing system focuses said excitation radiation on said surface of said substrate in a spot having a diameter of about 3 μm.
7. The system of claim 1, wherein said scanning field diameter is greater than about 10 mm.
8. The system of claim 1, wherein said scanning field diameter is about 14 mm.
9. The system of claim 1, wherein said numerical aperture is greater than about 0.25.
10. The system of claim 1, wherein said focusing system is achromatic.
11. The system of claim 1, wherein said radiation direction system is capable of scanning a spot across the substrate at a rate of at least 10 image lines/second.
12. The system of claim 1, wherein said radiation direction system is capable of scanning a spot across the substrate at a rate of at least 30 image lines/second.
13. The system of claim 1, wherein said radiation direction system comprises an angularly oscillating mirror or a rotating polyhedral mirror.
14. The system of claim 1, further comprising a translation stage upon which said substrate is mounted, said translation stage being moveable in at least one dimension perpendicular to an optical axis of said objective lens.
15. The system of claim 1, further comprising an autofocus system for placing said surface of said substrate in a focal plane of said focusing system.
16. The system of claim 1, further comprising a translation stage upon which said substrate is mounted, said translation stage being moveable in at least one dimension parallel to an optical axis of said focusing system and at least one dimension perpendicular to said optical axis of said focusing system.
17. The system of claim 1 further comprising a collection system comprising: collection optics for collecting fluorescence emitted from said surface of said substrate and for collecting an excitation radiation reflected from said surface of said substrate; separation optics for separating said emitted fluorescence from an excitation radiation reflected from said surface of said substrate and focusing said fluorescence through a confocal pinhole; and a recorder responsive to said fluorescence, for recording an amount of said fluorescence focused through said confocal pinhole.
18. The system of claim 1, wherein said surface of said substrate comprises a plurality of distinct polymer sequences in different known locations on said surface.
19. The system of claim 18, wherein each of said different polymer sequences is contained in a feature having at least one of a width or length dimension of less than about 50 μm.
20. The system of claim 1 further comprising a processor for processing and storing said signal so as to generate a 2-dimensional image of said sample.
21. The system of claim 1 further comprising a body for immobilizing said substrate, the substrate having at least a first surface with a sample thereon, the body comprising: a mounting surface; a cavity in said mounting surface, said first surface mated to said mounting surface for sealing said cavity, said sample being in fluid communication with said cavity, said cavity having a bottom surface comprising a light absorptive material; an inlet and an outlet being in communication with said cavity such that fluid flowing into said cavity for contacting said sample flows through said inlet and fluid flowing out of said cavity flows through said outlet; and a temperature controller for controlling the temperature in said cavity.
22. The system of claim 21 wherein said temperature controller comprises a thermoelectric cooler.
23. A system for detecting marked regions on a surface of a substrate, the system comprising: an excitation radiation source; first focusing optics for focusing said excitation radiation on said surface of said substrate in a spot having a diameter no greater than 5 μm, said focusing optics including an objective lens; a reciprocating radiation direction system for scanning said spot linearly across said surface of said substrate, said spot having a travel distance of at least 10 mm; an optical train for separating emissions from said surface of said substrate from excitation radiation reflected from said surface of said substrate, said optical train including said objective lens; and an autofocus system for automatically placing said surface of said substrate in a focal plane of said first focusing optics.
24. The system of claim 23, wherein said optical train includes a dichroic beam splitter for separating fluorescence emitted from said surface of said substrate from excitation radiation reflected from said surface of said substrate.
25. The system of claim 23, wherein said autofocus system comprises: second focusing optics for focusing said excitation radiation reflected from said surface of said substrate through a confocal pinhole onto a photodiode; and a translation stage upon which a substrate is mounted, said stage being capable of moving in a direction parallel with an optical axis of said first focusing optics for moving said substrate into a focal plane of said first focusing optics.
26. The system of claim 23 further comprising a detector for detecting an emission from said surface of said substrate in response to said excitation radiation, and a data acquisition system for recording an amount of said emission detected as a function of a position on said surface of said substrate from which said emission was emitted.
27. A method of scanning a polymer array having a plurality of different polymer sequences, each of said different polymer sequences being immobilized on a surface of a substrate in a different known location, to identify which polymer sequence on said array is bound by a target molecule, the method comprising: focusing an excitation radiation source upon said surface of said substrate with an objective lens; scanning said excitation radiation across said surface of said substrate at a speed of at least about 5 image lines/second; collecting emissions from said surface of said substrate in response to said excitation radiation with said objection lens; recording said emissions as a function of a position on said surface of said substrate, said position indicating the polymer sequence on said array is bound by a target molecule.
28. The method of claim 27, wherein said polymer array comprises a plurality of different oligonucleotide sequences immobilized on said surface of said substrate in different known locations.
29. The method of claim 27 further comprising focusing the excitation radiation source to a spot having a diameter of about 3 μm and scanning the spot in linear direction across the substrate for at least 14 mm.
30. The method of claim 27 further comprising the steps of: immobilizing said substrate on a body; exciting said sample on said substrate with an excitation radiation having a first wavelength from an electromagnetic radiation source, said excitation radiation detecting a response radiation having a second wavelength in response to said excitation radiation, said response radiation representing an image of said plurality of regions; exciting a subsequent plurality of regions on said sample; processing and storing said response radiation to generate a 2-dimensional image of said sample; and auto-focusing said sample in a focal plane of said excitation radiation.
31. The method as recited in claim 30 wherein said body comprises a mounting surface having a cavity thereon, said substrate being immobilized on said mounting surface such that said sample is in fluid communication with said reaction chamber, said reaction chamber comprising a inlet and a outlet for flowing fluids into and through said reaction chamber.
32. The method as recited in claim 31 wherein said body further comprises a temperature controller for controlling the temperature in said cavity.
33. The method as recited in claim 27 wherein said excitation radiation is scanned across said surface of said substrate at a speed of at least about 10 image lines/second.
34. The method as recited in claim 27 wherein said excitation radiation is scanned across said surface of said substrate at a speed of at least about 30 image lines/second.
PCT/US1997/008319 1996-05-16 1997-05-15 Systems and methods for detection of labeled materials WO1997043611A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
EP97925604A EP0902885A4 (en) 1996-05-16 1997-05-15 Systems and methods for detection of labeled materials
JP09541141A JP2000512744A (en) 1996-05-16 1997-05-15 System and method for detecting label material

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US1720396P 1996-05-16 1996-05-16
US60/017,203 1996-05-16

Publications (1)

Publication Number Publication Date
WO1997043611A1 true WO1997043611A1 (en) 1997-11-20

Family

ID=21781308

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1997/008319 WO1997043611A1 (en) 1996-05-16 1997-05-15 Systems and methods for detection of labeled materials

Country Status (4)

Country Link
US (4) US5981956A (en)
EP (1) EP0902885A4 (en)
JP (2) JP2000512744A (en)
WO (1) WO1997043611A1 (en)

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999052625A2 (en) * 1998-04-09 1999-10-21 Institut für Diagnostikforschung GmbH an der Freien Universität Berlin Method and device for producing synthesis or analysis series
WO1999063328A1 (en) * 1998-06-01 1999-12-09 Abbott Laboratories Sensor utilizing raman spectroscopy for non-invasive monitoring of analytes in biological fluid and method of use
WO2000063678A1 (en) * 1999-04-14 2000-10-26 Carl Zeiss Jena Gmbh Device for examining samples
JP2001091463A (en) * 1999-09-22 2001-04-06 Tosoh Corp Scanner type fluorescence detector using small-sized exciting light source
EP1311824A1 (en) * 2000-06-25 2003-05-21 Affymetrix, Inc. Optically active substrates
US6602714B1 (en) 1999-11-09 2003-08-05 Sri International Viscosity and mass sensor for the high-throughput synthesis, screening and characterization of combinatorial libraries
DE10236433A1 (en) * 2002-08-06 2004-02-19 Förderverein Institut für Medizintechnik Dresden e.V. Device for optically detecting liquid samples in droplet form formed discretely on planar substrate or in cavities of substrate comprises optical detector arranged in optical axis of light beam, and optical element
US6750963B2 (en) 2002-05-21 2004-06-15 Agilent Technologies, Inc. Imaging systems for signals on a surface
US7033840B1 (en) 1999-11-09 2006-04-25 Sri International Reaction calorimeter and differential scanning calorimeter for the high-throughput synthesis, screening and characterization of combinatorial libraries
EP2291539A1 (en) * 2008-06-25 2011-03-09 Real-Time Genomics, Llc Method and apparatus for melting curve analysis of nucleic acids in microarray format
USRE43097E1 (en) 1994-10-13 2012-01-10 Illumina, Inc. Massively parallel signature sequencing by ligation of encoded adaptors
CN104568873A (en) * 2014-12-22 2015-04-29 中国科学院苏州生物医学工程技术研究所 Laser scanning confocal microscope for imaging fluorescent substances
US9273354B2 (en) 1997-05-23 2016-03-01 Illumina, Inc. System and apparatus for sequential processing of analytes
WO2018033523A1 (en) * 2016-08-16 2018-02-22 Technische Universität Hamburg-Harburg Application profile reactor for operando measurements
CN108885260A (en) * 2016-04-08 2018-11-23 苹果公司 Transition time detector with single shaft scanning

Families Citing this family (295)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CH697814B1 (en) * 2001-01-26 2009-02-27 Tecan Trading Ag Optical system and method for exciting and measuring fluorescence on or in samples treated with fluorescent dyes.
US6955915B2 (en) * 1989-06-07 2005-10-18 Affymetrix, Inc. Apparatus comprising polymers
US20030017081A1 (en) * 1994-02-10 2003-01-23 Affymetrix, Inc. Method and apparatus for imaging a sample on a device
US5631734A (en) 1994-02-10 1997-05-20 Affymetrix, Inc. Method and apparatus for detection of fluorescently labeled materials
US6090555A (en) * 1997-12-11 2000-07-18 Affymetrix, Inc. Scanned image alignment systems and methods
US6741344B1 (en) * 1994-02-10 2004-05-25 Affymetrix, Inc. Method and apparatus for detection of fluorescently labeled materials
WO1997043611A1 (en) 1996-05-16 1997-11-20 Affymetrix, Inc. Systems and methods for detection of labeled materials
KR100618502B1 (en) * 1998-03-16 2006-09-01 지이 헬스케어 바이오-사이언시즈 코프. System and method of focusing electromagnetic radiation for use in a confocal microscopy imaging system
US6185030B1 (en) 1998-03-20 2001-02-06 James W. Overbeck Wide field of view and high speed scanning microscopy
US7875440B2 (en) 1998-05-01 2011-01-25 Arizona Board Of Regents Method of determining the nucleotide sequence of oligonucleotides and DNA molecules
US6780591B2 (en) 1998-05-01 2004-08-24 Arizona Board Of Regents Method of determining the nucleotide sequence of oligonucleotides and DNA molecules
US6472671B1 (en) 2000-02-09 2002-10-29 Jean I. Montagu Quantified fluorescence microscopy
US6342397B1 (en) * 1998-06-04 2002-01-29 Erkki Soini Homogeneous biospecific assay using a solid phase, two-photon excitation and confocal fluorescence detection
WO2000006996A1 (en) * 1998-07-28 2000-02-10 Ce Resources Pte Ltd. Optical detection system
US6486957B1 (en) * 1998-09-15 2002-11-26 Genesys Technologies, Inc. Replaceable filter module
CA2350692A1 (en) * 1998-10-29 2000-05-11 Cell Works Inc. Multiple marker characterization of single cells
WO2001015193A1 (en) * 1999-08-26 2001-03-01 Affymetrix, Inc. Systems and methods for high performance scanning
US6545264B1 (en) * 1998-10-30 2003-04-08 Affymetrix, Inc. Systems and methods for high performance scanning
US6824866B1 (en) 1999-04-08 2004-11-30 Affymetrix, Inc. Porous silica substrates for polymer synthesis and assays
US20060275782A1 (en) 1999-04-20 2006-12-07 Illumina, Inc. Detection of nucleic acid reactions on bead arrays
US7410793B2 (en) 1999-05-17 2008-08-12 Applera Corporation Optical instrument including excitation source
US20050279949A1 (en) * 1999-05-17 2005-12-22 Applera Corporation Temperature control for light-emitting diode stabilization
US7387891B2 (en) * 1999-05-17 2008-06-17 Applera Corporation Optical instrument including excitation source
US7501245B2 (en) * 1999-06-28 2009-03-10 Helicos Biosciences Corp. Methods and apparatuses for analyzing polynucleotide sequences
US6818395B1 (en) 1999-06-28 2004-11-16 California Institute Of Technology Methods and apparatus for analyzing polynucleotide sequences
DE60030978T2 (en) 1999-07-05 2007-06-14 Novartis Ag METHOD FOR USING A SENSOR UNIT
US6771376B2 (en) * 1999-07-05 2004-08-03 Novartis Ag Sensor platform, apparatus incorporating the platform, and process using the platform
US7217573B1 (en) * 1999-10-05 2007-05-15 Hitachi, Ltd. Method of inspecting a DNA chip
US6958225B2 (en) 1999-10-27 2005-10-25 Affymetrix, Inc. Complexity management of genomic DNA
US7167615B1 (en) 1999-11-05 2007-01-23 Board Of Regents, The University Of Texas System Resonant waveguide-grating filters and sensors and methods for making and using same
US6596483B1 (en) 1999-11-12 2003-07-22 Motorola, Inc. System and method for detecting molecules using an active pixel sensor
US6642046B1 (en) * 1999-12-09 2003-11-04 Motorola, Inc. Method and apparatus for performing biological reactions on a substrate surface
US6884578B2 (en) * 2000-03-31 2005-04-26 Affymetrix, Inc. Genes differentially expressed in secretory versus proliferative endometrium
US6586750B2 (en) 2000-08-03 2003-07-01 Perlegen Sciences High performance substrate scanning
DE10038080A1 (en) * 2000-08-04 2002-02-21 Giesing Michael Registering the presence of immobilized substances on a bio-chip carrier, comprises using a fluorescence scanner, where a pulsed laser excites fluorescent markings to be detected between the pulses with local resolution
US6545758B1 (en) * 2000-08-17 2003-04-08 Perry Sandstrom Microarray detector and synthesizer
US6567163B1 (en) 2000-08-17 2003-05-20 Able Signal Company Llc Microarray detector and synthesizer
CA2388064A1 (en) 2000-08-22 2002-05-02 Affymetrix, Inc. System method, and computer software product for controlling biological microarray scanner
US6789040B2 (en) 2000-08-22 2004-09-07 Affymetrix, Inc. System, method, and computer software product for specifying a scanning area of a substrate
US7108969B1 (en) 2000-09-08 2006-09-19 Affymetrix, Inc. Methods for detecting and diagnosing oral cancer
US6674058B1 (en) * 2000-09-20 2004-01-06 Compucyte Corporation Apparatus and method for focusing a laser scanning cytometer
EP1203945B1 (en) * 2000-10-26 2006-12-20 Agilent Technologies, Inc. (a Delaware corporation) Microarray
US6832163B2 (en) * 2001-02-09 2004-12-14 Agilent Technologies, Inc. Methods of identifying heterogeneous features in an image of an array
EP2089413A4 (en) * 2001-02-21 2009-10-28 Amnis Corp Method and apparatus for labeling and analyzing cellular components
EP2295971B1 (en) * 2001-03-09 2016-09-07 TrovaGene, Inc. Conjugate probes and optical detection of analytes
JP2004523243A (en) * 2001-03-12 2004-08-05 カリフォルニア インスティチュート オブ テクノロジー Method and apparatus for analyzing polynucleotide sequences by asynchronous base extension
KR100396954B1 (en) * 2001-03-27 2003-09-03 임채헌 Fiter box for analysing wave length of light and cell analysis device utilizing the same
AU785425B2 (en) * 2001-03-30 2007-05-17 Genetic Technologies Limited Methods of genomic analysis
US7115726B2 (en) * 2001-03-30 2006-10-03 Perlegen Sciences, Inc. Haplotype structures of chromosome 21
US6650411B2 (en) 2001-04-26 2003-11-18 Affymetrix, Inc. System, method, and product for pixel clocking in scanning of biological materials
US6490533B2 (en) * 2001-04-26 2002-12-03 Affymetrix, Inc. System, method, and product for dynamic noise reduction in scanning of biological materials
US6643015B2 (en) 2001-04-26 2003-11-04 Affymetrix, Inc. System, method, and product for symmetrical filtering in scanning of biological materials
DE10121064A1 (en) * 2001-04-28 2002-10-31 Evotec Ag Device and method for the optical measurement of chemical and / or biological samples
US20040238730A1 (en) * 2001-05-07 2004-12-02 Jorg Langowski Fluorescence fluctuation microscope analytical module or scanning module, method for measurement of fluorescence fluctuation and method and device for adjustment of a fluorescence fluctuation microscope
US6662091B2 (en) * 2001-06-29 2003-12-09 Battelle Memorial Institute Diagnostics/prognostics using wireless links
US20080026367A9 (en) * 2001-08-17 2008-01-31 Perlegen Sciences, Inc. Methods for genomic analysis
US20060188875A1 (en) * 2001-09-18 2006-08-24 Perlegen Sciences, Inc. Human genomic polymorphisms
US20030068621A1 (en) * 2001-10-04 2003-04-10 Jonathan Briggs Method and device for producing oligonucleotide arrays
US20040053232A1 (en) * 2001-10-05 2004-03-18 Perlegen Sciences, Inc. Haplotype structures of chromosome 21
ITMI20010566U1 (en) * 2001-10-25 2003-04-25 Troletti Adriano HAIR DRYER, WITH HIGH PRACTICALITY OF USE
DE10155600B4 (en) * 2001-11-09 2009-08-27 Oligene Gmbh Nucleic acid array
DE10157511A1 (en) * 2001-11-23 2003-06-12 Evotec Ag Method and device for correcting the size and / or shape of a measurement volume in a chemical and / or biological sample
US20040023237A1 (en) * 2001-11-26 2004-02-05 Perelegen Sciences Inc. Methods for genomic analysis
EP1316794A1 (en) * 2001-11-28 2003-06-04 Cambridge University Technical Services Limited A dual wavelength optical fluorescence analyser
US7635588B2 (en) * 2001-11-29 2009-12-22 Applied Biosystems, Llc Apparatus and method for differentiating multiple fluorescence signals by excitation wavelength
US6794424B2 (en) * 2001-12-04 2004-09-21 Agilent Technologies, Inc. Devices for calibrating optical scanners and methods of using the same
CN1281324C (en) * 2001-12-19 2006-10-25 阿菲梅特里克斯公司 Manufacturing process for array plate assembly
US6770892B2 (en) * 2002-02-28 2004-08-03 Agilent Technologies, Inc. Method and system for automated focus-distance determination for molecular array scanners
EP1345026B1 (en) 2002-03-15 2010-05-05 Affymetrix, Inc. System and method for scanning of biological materials
US20040023275A1 (en) * 2002-04-29 2004-02-05 Perlegen Sciences, Inc. Methods for genomic analysis
US6658143B2 (en) * 2002-04-29 2003-12-02 Amersham Biosciences Corp. Ray-based image analysis for biological specimens
US6791690B2 (en) * 2002-04-30 2004-09-14 Agilent Technologies, Inc. Reading dry chemical arrays
JP3729154B2 (en) * 2002-05-10 2005-12-21 株式会社日立製作所 Pattern defect inspection method and apparatus
JP2005526253A (en) 2002-05-17 2005-09-02 アプレラ コーポレイション Apparatus and method for differentiating multiple fluorescent signals by excitation wavelength
AU2003249681A1 (en) * 2002-05-31 2003-12-19 Diversa Corporation Multiplexed systems for nucleic acid sequencing
JP4377811B2 (en) * 2002-06-21 2009-12-02 オリンパス株式会社 Biomolecule analyzer and biomolecule analysis method
US7504215B2 (en) 2002-07-12 2009-03-17 Affymetrix, Inc. Nucleic acid labeling methods
DE10234524A1 (en) * 2002-07-24 2004-02-12 Oligene Gmbh Array of probes derived from monocyte-macrophage genes, useful e.g. for diagnosis, prognosis and therapeutic monitoring of rheumatoid arthritis and other inflammatory diseases
US7452712B2 (en) * 2002-07-30 2008-11-18 Applied Biosystems Inc. Sample block apparatus and method of maintaining a microcard on a sample block
US20040038214A1 (en) * 2002-08-23 2004-02-26 Corson John F. Method and system for reading a molecular array
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
US6970240B2 (en) * 2003-03-10 2005-11-29 Applera Corporation Combination reader
US7534561B2 (en) 2003-04-02 2009-05-19 Agilent Technologies, Inc. Nucleic acid array in situ fabrication methods and arrays produced using the same
EP1477258A1 (en) * 2003-05-16 2004-11-17 Fisba Optik Ag Device and method for local temperature treatment with heat detector and image treatment
EP3305919A1 (en) 2003-06-10 2018-04-11 The Trustees of Boston University Detection methods for disorders of the lung
US20050170367A1 (en) * 2003-06-10 2005-08-04 Quake Stephen R. Fluorescently labeled nucleoside triphosphates and analogs thereof for sequencing nucleic acids
US20040259270A1 (en) * 2003-06-19 2004-12-23 Wolf David E. System, device and method for exciting a sensor and detecting analyte
US20040259100A1 (en) 2003-06-20 2004-12-23 Illumina, Inc. Methods and compositions for whole genome amplification and genotyping
KR20060030510A (en) * 2003-07-11 2006-04-10 코니카 미놀타 옵토 인코포레이티드 Optical pickup device, optical element used for optical pickup device, and method for producing optical element
US7317415B2 (en) 2003-08-08 2008-01-08 Affymetrix, Inc. System, method, and product for scanning of biological materials employing dual analog integrators
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
CA2537254A1 (en) 2003-09-03 2005-03-17 Government Of The United States Of America, As Represented By Secretary, Department Of Health And Human Services Methods for identifying, diagnosing, and predicting survival of lymphomas
US7169560B2 (en) 2003-11-12 2007-01-30 Helicos Biosciences Corporation Short cycle methods for sequencing polynucleotides
US7304310B1 (en) 2003-11-21 2007-12-04 Kla-Tencor Technologies Corp. Methods and systems for inspecting a specimen using light scattered in different wavelength ranges
US7017812B1 (en) 2003-11-26 2006-03-28 The United States Of America As Represented By The Administrator Of The National Aeronautics And Space Administration Variable distance angular symbology reader
JP4790621B2 (en) 2003-11-26 2011-10-12 アドバンディーエックス, インコーポレイテッド Peptide nucleic acid probes for analysis of specific Staphylococcus species
US20060172408A1 (en) * 2003-12-01 2006-08-03 Quake Steven R Device for immobilizing chemical and biochemical species and methods of using same
CA2497324A1 (en) 2004-02-17 2005-08-17 Affymetrix, Inc. Methods for fragmenting and labelling dna
US7981604B2 (en) 2004-02-19 2011-07-19 California Institute Of Technology Methods and kits for analyzing polynucleotide sequences
WO2005082110A2 (en) * 2004-02-26 2005-09-09 Illumina Inc. Haplotype markers for diagnosing susceptibility to immunological conditions
US20060046258A1 (en) * 2004-02-27 2006-03-02 Lapidus Stanley N Applications of single molecule sequencing
US20050239085A1 (en) * 2004-04-23 2005-10-27 Buzby Philip R Methods for nucleic acid sequence determination
US20050260609A1 (en) * 2004-05-24 2005-11-24 Lapidus Stanley N Methods and devices for sequencing nucleic acids
EP1766090B1 (en) * 2004-05-25 2011-04-27 Helicos Biosciences Corporation Methods for nucleic acid immobilisation
US20070117103A1 (en) * 2005-11-22 2007-05-24 Buzby Philip R Nucleotide analogs
US20070117104A1 (en) * 2005-11-22 2007-05-24 Buzby Philip R Nucleotide analogs
US7476734B2 (en) * 2005-12-06 2009-01-13 Helicos Biosciences Corporation Nucleotide analogs
US7276720B2 (en) 2004-07-19 2007-10-02 Helicos Biosciences Corporation Apparatus and methods for analyzing samples
AU2005264061B2 (en) * 2004-07-23 2009-05-21 Ge Healthcare Niagara, Inc. Method and apparatus for fluorescent confocal microscopy
US20060024678A1 (en) * 2004-07-28 2006-02-02 Helicos Biosciences Corporation Use of single-stranded nucleic acid binding proteins in sequencing
US20060073506A1 (en) 2004-09-17 2006-04-06 Affymetrix, Inc. Methods for identifying biological samples
ATE492650T1 (en) * 2004-10-01 2011-01-15 Koninkl Philips Electronics Nv METHOD AND DEVICE FOR DETECTING MARKING ELEMENTS IN A SAMPLE
US20060073511A1 (en) 2004-10-05 2006-04-06 Affymetrix, Inc. Methods for amplifying and analyzing nucleic acids
JP2006126204A (en) 2004-10-29 2006-05-18 Affymetrix Inc Automated method for manufacturing polymer array
US7682782B2 (en) 2004-10-29 2010-03-23 Affymetrix, Inc. System, method, and product for multiple wavelength detection using single source excitation
US7647186B2 (en) * 2004-12-07 2010-01-12 Illumina, Inc. Oligonucleotide ordering system
US20060118754A1 (en) * 2004-12-08 2006-06-08 Lapen Daniel C Stabilizing a polyelectrolyte multilayer
US7220549B2 (en) * 2004-12-30 2007-05-22 Helicos Biosciences Corporation Stabilizing a nucleic acid for nucleic acid sequencing
US20060172328A1 (en) * 2005-01-05 2006-08-03 Buzby Philip R Methods and compositions for correcting misincorporation in a nucleic acid synthesis reaction
EP1681556B1 (en) * 2005-01-18 2007-04-11 Roche Diagnostics GmbH Imaging fluorescence signals using telecentricity
US20060166224A1 (en) * 2005-01-24 2006-07-27 Norviel Vernon A Associations using genotypes and phenotypes
US7482120B2 (en) * 2005-01-28 2009-01-27 Helicos Biosciences Corporation Methods and compositions for improving fidelity in a nucleic acid synthesis reaction
US7274445B1 (en) * 2005-03-11 2007-09-25 Kla-Tencor Technologies Corporation Confocal scatterometer and method for single-sided detection of particles and defects on a transparent wafer or disk
US7768638B2 (en) 2005-03-18 2010-08-03 Illumina, Inc. Systems for and methods of facilitating focusing an optical scanner
AU2006236621B2 (en) 2005-04-14 2011-10-06 The Trustees Of Boston University Diagnostic for lung disorders using class prediction
US8351026B2 (en) 2005-04-22 2013-01-08 Affymetrix, Inc. Methods and devices for reading microarrays
US20060263790A1 (en) * 2005-05-20 2006-11-23 Timothy Harris Methods for improving fidelity in a nucleic acid synthesis reaction
DK2463386T3 (en) 2005-06-15 2017-07-31 Complete Genomics Inc Nucleic acid analysis using random mixtures of non-overlapping fragments
US7397042B2 (en) * 2005-08-24 2008-07-08 Dr. Chip Biotechnology Incorporation Optical detection apparatus and method thereof
US7666593B2 (en) 2005-08-26 2010-02-23 Helicos Biosciences Corporation Single molecule sequencing of captured nucleic acids
AU2006315664B2 (en) * 2005-11-10 2012-03-08 Tautheta Instruments Llc Apparatus and method for system identification
US20070117102A1 (en) * 2005-11-22 2007-05-24 Buzby Philip R Nucleotide analogs
US7329860B2 (en) * 2005-11-23 2008-02-12 Illumina, Inc. Confocal imaging methods and apparatus
US20070128610A1 (en) * 2005-12-02 2007-06-07 Buzby Philip R Sample preparation method and apparatus for nucleic acid sequencing
US7634363B2 (en) * 2005-12-07 2009-12-15 Affymetrix, Inc. Methods for high throughput genotyping
US20070161031A1 (en) * 2005-12-16 2007-07-12 The Board Of Trustees Of The Leland Stanford Junior University Functional arrays for high throughput characterization of gene expression regulatory elements
US8055098B2 (en) 2006-01-27 2011-11-08 Affymetrix, Inc. System, method, and product for imaging probe arrays with small feature sizes
US9445025B2 (en) 2006-01-27 2016-09-13 Affymetrix, Inc. System, method, and product for imaging probe arrays with small feature sizes
US20090080194A1 (en) * 2006-02-15 2009-03-26 Li-Cor, Inc. Fluorescence filtering system and method for molecular imaging
JP2009529329A (en) 2006-03-09 2009-08-20 トラスティーズ オブ ボストン ユニバーシティ Methods for diagnosis and prognosis for lung diseases using gene expression profiles of nasal epithelial cells
US7914988B1 (en) 2006-03-31 2011-03-29 Illumina, Inc. Gene expression profiles to predict relapse of prostate cancer
DE102006019468B3 (en) * 2006-04-26 2007-08-30 Siemens Ag Optical sensor for collection of point shaped, linear shaped or laminar defects at plane surface area, has telecentric laser scanner comprises laser for approximately perpendicular lighting of flat surface area and has scanning reflector
US20100216657A1 (en) * 2006-05-16 2010-08-26 Arcxis Biotechnologies, Inc. Pcr-free sample preparation and detection systems for high speed biologic analysis and identification
US11001881B2 (en) * 2006-08-24 2021-05-11 California Institute Of Technology Methods for detecting analytes
US11098345B2 (en) * 2006-06-05 2021-08-24 California Institute Of Technology Methods for detecting target analytes
US8009889B2 (en) 2006-06-27 2011-08-30 Affymetrix, Inc. Feature intensity reconstruction of biological probe array
KR100808762B1 (en) 2006-07-06 2008-02-29 충북대학교 산학협력단 Detecting Method For Nano Bio-Chip
WO2008010120A2 (en) * 2006-07-17 2008-01-24 Koninklijke Philips Electronics N.V. Employing beam scanning for optical detection
US8048626B2 (en) 2006-07-28 2011-11-01 California Institute Of Technology Multiplex Q-PCR arrays
US11525156B2 (en) 2006-07-28 2022-12-13 California Institute Of Technology Multiplex Q-PCR arrays
EP2057465A4 (en) 2006-08-09 2010-04-21 Homestead Clinical Corp Organ-specific proteins and methods of their use
US11560588B2 (en) 2006-08-24 2023-01-24 California Institute Of Technology Multiplex Q-PCR arrays
JP5473202B2 (en) * 2006-10-13 2014-04-16 滋賀県 Method and system for detecting fluorescent material in a sample
US9845494B2 (en) 2006-10-18 2017-12-19 Affymetrix, Inc. Enzymatic methods for genotyping on arrays
WO2008066655A2 (en) 2006-11-02 2008-06-05 Yale University Assessment of oocyte competence
US7791013B2 (en) * 2006-11-21 2010-09-07 Illumina, Inc. Biological microarray line scanning method and system
US7813013B2 (en) * 2006-11-21 2010-10-12 Illumina, Inc. Hexagonal site line scanning method and system
US8293684B2 (en) * 2006-11-29 2012-10-23 Exiqon Locked nucleic acid reagents for labelling nucleic acids
US7817275B2 (en) * 2007-03-07 2010-10-19 University Of Delaware Scanning optical microscope with long working distance objective
WO2008112127A2 (en) * 2007-03-08 2008-09-18 Switchgear Genomics Functional arrays for high throughput characterization of regulatory elements in untranslated regions of genes
ES2748134T3 (en) * 2007-04-04 2020-03-13 Ande Corp Systems for rapid multiple amplification of target nucleic acids
EP2076289B1 (en) 2007-04-13 2014-11-12 Dana-Farber Cancer Institute, Inc. Methods for treating cancer resistant to erbb therapeutics
WO2008144600A1 (en) * 2007-05-17 2008-11-27 Prescient Medical, Inc. Multi-channel fiber optic spectroscopy systems employing integrated optics modules
US8200440B2 (en) 2007-05-18 2012-06-12 Affymetrix, Inc. System, method, and computer software product for genotype determination using probe array data
US7952719B2 (en) * 2007-06-08 2011-05-31 Prescient Medical, Inc. Optical catheter configurations combining raman spectroscopy with optical fiber-based low coherence reflectometry
US8629382B2 (en) * 2007-09-03 2014-01-14 Nikon Corporation Auto focus apparatus for detecting a focal point with a setting section for shifting an irradiation position outside an observation field of an imaging section
US20090112482A1 (en) * 2007-10-26 2009-04-30 Sandstrom Perry L Microarray detector and synthesizer
DE102007052517A1 (en) 2007-10-29 2009-04-30 Autoimmun Diagnostika Gmbh ELISPOT process with two filter systems
CN101868713A (en) * 2007-11-23 2010-10-20 皇家飞利浦电子股份有限公司 Beam shaper, optical system and using method thereof
KR100922577B1 (en) * 2007-12-13 2009-10-23 한국전자통신연구원 Portable biophotonic sensor measurement system
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
CA2710807C (en) 2008-03-11 2015-09-08 Kyeong Man Hong Method for measuring chromosome, gene or specific nucleotide sequence copy numbers using snp array
US8134698B1 (en) 2008-03-14 2012-03-13 Kla-Tencor Corporation Dynamic range extension in surface inspection systems
US11035823B2 (en) * 2009-03-17 2021-06-15 Qiagen Sciences, Llc Methods and devices for sequencing nucleic acids in smaller batches
US8628940B2 (en) 2008-09-24 2014-01-14 Pacific Biosciences Of California, Inc. Intermittent detection during analytical reactions
MX2010010600A (en) 2008-03-28 2011-03-30 Pacific Biosciences California Inc Compositions and methods for nucleic acid sequencing.
US8143030B2 (en) 2008-09-24 2012-03-27 Pacific Biosciences Of California, Inc. Intermittent detection during analytical reactions
US8236499B2 (en) 2008-03-28 2012-08-07 Pacific Biosciences Of California, Inc. Methods and compositions for nucleic acid sample preparation
US8039817B2 (en) 2008-05-05 2011-10-18 Illumina, Inc. Compensator for multiple surface imaging
WO2009137521A2 (en) * 2008-05-07 2009-11-12 Illumina, Inc. Compositions and methods for providing substances to and from an array
US20110195064A1 (en) 2008-06-06 2011-08-11 The United States Of America, As Represented By The Secretary, Department Of Health And Human Serv Survival predictor for diffuse large b cell lymphoma
US8383369B2 (en) 2008-09-24 2013-02-26 Pacific Biosciences Of California, Inc. Intermittent detection during analytical reactions
US20100087325A1 (en) * 2008-10-07 2010-04-08 Illumina, Inc. Biological sample temperature control system and method
US8541207B2 (en) 2008-10-22 2013-09-24 Illumina, Inc. Preservation of information related to genomic DNA methylation
US20100113906A1 (en) * 2008-11-06 2010-05-06 Prescient Medical, Inc. Hybrid basket catheters
US20100157086A1 (en) * 2008-12-15 2010-06-24 Illumina, Inc Dynamic autofocus method and system for assay imager
US8374818B2 (en) * 2008-12-19 2013-02-12 Affymetrix, Inc. System, method and apparatus for calibrating inspection tools
JP2010148057A (en) * 2008-12-22 2010-07-01 Sharp Corp Power amplifier, integrated circuit, and communication apparatus
WO2010091406A2 (en) * 2009-02-09 2010-08-12 Forensic Science Service Limited Improvements in and relating to devices
CN102439454B (en) 2009-02-11 2015-05-13 卡里斯Mpi公司 Molecular profiling of tumors
EP2409139A4 (en) * 2009-03-18 2016-10-12 Univ Utah Res Found Non-coherent light microscopy
WO2010108038A2 (en) 2009-03-18 2010-09-23 University Of Utah Research Foundation A microscopy system and method for creating three dimensional images using probe molecules
JP5167194B2 (en) * 2009-04-28 2013-03-21 株式会社日立エンジニアリング・アンド・サービス Microbiological testing device
US9767342B2 (en) 2009-05-22 2017-09-19 Affymetrix, Inc. Methods and devices for reading microarrays
US10174368B2 (en) 2009-09-10 2019-01-08 Centrillion Technology Holdings Corporation Methods and systems for sequencing long nucleic acids
WO2011032040A1 (en) 2009-09-10 2011-03-17 Centrillion Technology Holding Corporation Methods of targeted sequencing
US8815779B2 (en) 2009-09-16 2014-08-26 SwitchGear Genomics, Inc. Transcription biomarkers of biological responses and methods
AU2010315400B2 (en) 2009-10-27 2016-07-21 Caris Mpi, Inc. Molecular profiling for personalized medicine
US8501122B2 (en) 2009-12-08 2013-08-06 Affymetrix, Inc. Manufacturing and processing polymer arrays
US8835358B2 (en) 2009-12-15 2014-09-16 Cellular Research, Inc. Digital counting of individual molecules by stochastic attachment of diverse labels
US8422031B2 (en) 2010-02-01 2013-04-16 Illumina, Inc. Focusing methods and optical systems and assemblies using the same
DE202011003570U1 (en) 2010-03-06 2012-01-30 Illumina, Inc. Systems and apparatus for detecting optical signals from a sample
WO2011159942A1 (en) 2010-06-18 2011-12-22 Illumina, Inc. Conformational probes and methods for sequencing nucleic acids
US11031095B2 (en) 2010-08-06 2021-06-08 Ariosa Diagnostics, Inc. Assay systems for determination of fetal copy number variation
US20130261003A1 (en) 2010-08-06 2013-10-03 Ariosa Diagnostics, In. Ligation-based detection of genetic variants
US8700338B2 (en) 2011-01-25 2014-04-15 Ariosa Diagnosis, Inc. Risk calculation for evaluation of fetal aneuploidy
US10533223B2 (en) 2010-08-06 2020-01-14 Ariosa Diagnostics, Inc. Detection of target nucleic acids using hybridization
US20120034603A1 (en) 2010-08-06 2012-02-09 Tandem Diagnostics, Inc. Ligation-based detection of genetic variants
US11203786B2 (en) 2010-08-06 2021-12-21 Ariosa Diagnostics, Inc. Detection of target nucleic acids using hybridization
US20130040375A1 (en) 2011-08-08 2013-02-14 Tandem Diagnotics, Inc. Assay systems for genetic analysis
US20140342940A1 (en) 2011-01-25 2014-11-20 Ariosa Diagnostics, Inc. Detection of Target Nucleic Acids using Hybridization
US10167508B2 (en) 2010-08-06 2019-01-01 Ariosa Diagnostics, Inc. Detection of genetic abnormalities
US9029103B2 (en) 2010-08-27 2015-05-12 Illumina Cambridge Limited Methods for sequencing polynucleotides
WO2012050920A1 (en) 2010-09-29 2012-04-19 Illumina, Inc. Compositions and methods for sequencing nucleic acids
WO2012055929A1 (en) 2010-10-26 2012-05-03 Illumina, Inc. Sequencing methods
US8575071B2 (en) 2010-11-03 2013-11-05 Illumina, Inc. Reducing adapter dimer formation
US9494783B2 (en) * 2010-11-30 2016-11-15 Etaluma Inc. Compact, high-resolution fluorescence and brightfield microscope and methods of use
US8951781B2 (en) 2011-01-10 2015-02-10 Illumina, Inc. Systems, methods, and apparatuses to image a sample for biological or chemical analysis
WO2012103031A2 (en) 2011-01-25 2012-08-02 Ariosa Diagnostics, Inc. Detection of genetic abnormalities
US9994897B2 (en) 2013-03-08 2018-06-12 Ariosa Diagnostics, Inc. Non-invasive fetal sex determination
US10131947B2 (en) 2011-01-25 2018-11-20 Ariosa Diagnostics, Inc. Noninvasive detection of fetal aneuploidy in egg donor pregnancies
US11270781B2 (en) 2011-01-25 2022-03-08 Ariosa Diagnostics, Inc. Statistical analysis for non-invasive sex chromosome aneuploidy determination
US8756020B2 (en) 2011-01-25 2014-06-17 Ariosa Diagnostics, Inc. Enhanced risk probabilities using biomolecule estimations
CA2827200A1 (en) 2011-02-24 2012-08-30 Hill's Pet Nutrition, Inc. Compositions and methods for diagnosing and treating kidney disorders in a feline
WO2012118745A1 (en) 2011-02-28 2012-09-07 Arnold Oliphant Assay systems for detection of aneuploidy and sex determination
US20120252682A1 (en) 2011-04-01 2012-10-04 Maples Corporate Services Limited Methods and systems for sequencing nucleic acids
EP2707509A4 (en) 2011-05-12 2014-11-26 Netbio Inc Methods and compositions for rapid multiplex amplification of str loci
EP2718465B1 (en) 2011-06-09 2022-04-13 Illumina, Inc. Method of making an analyte array
CN103597097B (en) 2011-06-15 2016-08-17 希尔氏宠物营养品公司 For diagnosing and monitor hyperthyroid compositions and the method for felid
US8712697B2 (en) 2011-09-07 2014-04-29 Ariosa Diagnostics, Inc. Determination of copy number variations using binomial probability calculations
US9599561B2 (en) 2011-10-13 2017-03-21 Affymetrix, Inc. Methods, systems and apparatuses for testing and calibrating fluorescent scanners
US9176293B2 (en) 2011-10-28 2015-11-03 Corning Cable Systems Llc Buffered fibers with access features
EP2771103B1 (en) 2011-10-28 2017-08-16 Illumina, Inc. Microarray fabrication system and method
RU2612901C2 (en) 2011-12-19 2017-03-13 Хилл'С Пет Ньютришн, Инк. Compositions and methods for diagnosing and treating hyperthyroidism of companion animals
WO2013117595A2 (en) 2012-02-07 2013-08-15 Illumina Cambridge Limited Targeted enrichment and amplification of nucleic acids on a support
WO2013123125A1 (en) 2012-02-17 2013-08-22 President And Fellows Of Harvard College Assembly of nucleic acid sequences in emulsions
CA2865575C (en) 2012-02-27 2024-01-16 Cellular Research, Inc. Compositions and kits for molecular counting
EP2820174B1 (en) 2012-02-27 2019-12-25 The University of North Carolina at Chapel Hill Methods and uses for molecular tags
JP5896797B2 (en) * 2012-03-16 2016-03-30 オリンパス株式会社 Pixel clock generator
US10289800B2 (en) 2012-05-21 2019-05-14 Ariosa Diagnostics, Inc. Processes for calculating phased fetal genomic sequences
WO2014015269A1 (en) 2012-07-19 2014-01-23 Ariosa Diagnostics, Inc. Multiplexed sequential ligation-based detection of genetic variants
CA2881823C (en) 2012-08-20 2019-06-11 Illumina, Inc. Method and system for fluorescence lifetime based sequencing
TWI619809B (en) * 2012-08-24 2018-04-01 佐竹股份有限公司 Method and device for inspecting microorganisms
EP2932266A4 (en) 2012-12-17 2016-11-30 Leukodx Ltd Systems and methods for determining a chemical state
US10610861B2 (en) 2012-12-17 2020-04-07 Accellix Ltd. Systems, compositions and methods for detecting a biological condition
US10838406B2 (en) 2013-02-11 2020-11-17 The Aerospace Corporation Systems and methods for the patterning of material substrates
US10613513B2 (en) * 2013-02-11 2020-04-07 The Aerospace Corporation Systems and methods for modifying material substrates
US9146248B2 (en) 2013-03-14 2015-09-29 Intelligent Bio-Systems, Inc. Apparatus and methods for purging flow cells in nucleic acid sequencing instruments
US10119134B2 (en) 2013-03-15 2018-11-06 Abvitro Llc Single cell bar-coding for antibody discovery
US9591268B2 (en) 2013-03-15 2017-03-07 Qiagen Waltham, Inc. Flow cell alignment methods and systems
ES2751402T3 (en) 2013-03-27 2020-03-31 Bluegnome Ltd Aneuploidy risk assessment
WO2014200579A1 (en) 2013-06-13 2014-12-18 Ariosa Diagnostics, Inc. Statistical analysis for non-invasive sex chromosome aneuploidy determination
SG10201806890VA (en) 2013-08-28 2018-09-27 Cellular Res Inc Massively parallel single cell analysis
DE102013022538B3 (en) * 2013-09-03 2018-12-13 Georg-August-Universität Göttingen Stiftung Öffentlichen Rechts Method for creating a microscope image and microscopy device
US9352315B2 (en) 2013-09-27 2016-05-31 Taiwan Semiconductor Manufacturing Company, Ltd. Method to produce chemical pattern in micro-fluidic structure
ES2738289T3 (en) 2013-11-06 2020-01-21 Hospital Clinic Barcelona Procedure for the subtyping of lymphoma types by means of expression profiles
JP2015125177A (en) * 2013-12-25 2015-07-06 ソニー株式会社 Microscope system, and image data transfer method
WO2015103225A1 (en) 2013-12-31 2015-07-09 Illumina, Inc. Addressable flow cell using patterned electrodes
AU2015206336B2 (en) 2014-01-16 2020-01-23 Illumina, Inc. Gene expression panel for prognosis of prostate cancer recurrence
DE102014000473A1 (en) * 2014-01-16 2015-07-16 Carl Zeiss Microscopy Gmbh Laser scanning microscope and amplifier module
JP6171970B2 (en) 2014-02-10 2017-08-02 ソニー株式会社 Laser scanning microscope apparatus and control method
SG11201608600YA (en) 2014-04-14 2016-11-29 Stanford Res Inst Int Portable nucleic acid analysis system and high-performance microfluidic electroactive polymer actuators
US9541750B2 (en) 2014-06-23 2017-01-10 Li-Cor, Inc. Telecentric, wide-field fluorescence scanning systems and methods
JP2016031234A (en) * 2014-07-25 2016-03-07 高電工業株式会社 Specimen checkup device
KR102541849B1 (en) 2014-09-15 2023-06-09 에이비비트로, 엘엘씨 High-throughput nucleotide library sequencing
WO2016130572A2 (en) 2015-02-10 2016-08-18 Dana-Farber Cancer Institute, Inc. Methods of determining levels of exposure to radiation and uses thereof
CA3077811C (en) 2015-03-24 2024-02-27 Illumina, Inc. Methods, carrier assemblies, and systems for imaging samples for biological or chemical analysis
KR102134953B1 (en) 2015-05-29 2020-07-16 일루미나, 인코포레이티드 Sample carrier and assay system for carrying out the specified reaction
CN106434873B (en) 2015-08-13 2021-08-27 生捷科技控股公司 Method for synchronizing nucleic acid molecules
CN108474805A (en) 2015-08-24 2018-08-31 亿明达股份有限公司 For accumulator and flow control system in biological and chemical setting-out line road
EP3353326B1 (en) 2015-09-24 2022-08-24 AbVitro LLC Affinity-oligonucleotide conjugates and uses thereof
WO2018057051A1 (en) 2016-09-24 2018-03-29 Abvitro Llc Affinity-oligonucleotide conjugates and uses thereof
JP7341661B2 (en) 2015-09-25 2023-09-11 アブビトロ, エルエルシー High-throughput process for T cell receptor target identification of natively paired T cell receptor sequences
IL296418A (en) 2015-10-18 2022-11-01 Affymetrix Inc Multiallelic genotyping of single nucleotide polymorphisms and indels
WO2017155858A1 (en) 2016-03-07 2017-09-14 Insilixa, Inc. Nucleic acid sequence identification using solid-phase cyclic single base extension
AU2017254645B2 (en) 2016-04-20 2023-02-09 Board Of Regents Of The University Of Nebraska Evaluation of mantle cell lymphoma and methods related thereto
WO2018064116A1 (en) 2016-09-28 2018-04-05 Illumina, Inc. Methods and systems for data compression
AU2016388716A1 (en) * 2016-12-22 2018-07-12 Advanced Optical Technologies, Inc. Polarimeter with multiple independent tunable channels and method for material and object classification and recognition
US11867891B2 (en) 2016-12-22 2024-01-09 Advanced Optical Technologies, Inc. Polarimeter with multiple independent tunable channels and method for material orientation imaging
FR3063932B1 (en) * 2017-03-15 2019-03-22 Universite de Bordeaux EQUIPMENT AND METHOD FOR PARTICLE DEPOSITION ON A TARGET
WO2018213803A1 (en) 2017-05-19 2018-11-22 Neon Therapeutics, Inc. Immunogenic neoantigen identification
EP3638814B8 (en) 2017-06-14 2023-06-21 The United States of America, as Represented by The Secretary, Department of Health and Human Services Method for determining lymphoma type
JP2022512080A (en) 2018-11-30 2022-02-02 カリス エムピーアイ インコーポレイテッド Next Generation Molecular Profiling
AU2019400090A1 (en) 2018-12-14 2021-01-07 Illumina Cambridge Limited Decreasing phasing with unlabeled nucleotides during sequencing
CA3103744A1 (en) 2018-12-17 2020-06-25 Pietro GATTI-LAFRANCONI Compositions for use in polynucleotide sequencing
CA3103750A1 (en) 2018-12-17 2020-06-25 Illumina Cambridge Limited Primer oligonucleotide for sequencing
JP2022525322A (en) 2019-03-14 2022-05-12 インシリクサ, インコーポレイテッド Methods and systems for time-gate fluorescence-based detection
BR112021020779A8 (en) 2019-04-17 2022-01-25 Igenomix S L Improved methods for early diagnosis of uterine leiomyomas and leiomyosarcomas
EP4069865A4 (en) 2019-12-02 2023-12-20 Caris MPI, Inc. Pan-cancer platinum response predictor
US20230096386A1 (en) 2021-09-30 2023-03-30 Illumina Cambridge Limited Polynucleotide sequencing
WO2023122363A1 (en) 2021-12-23 2023-06-29 Illumina Software, Inc. Dynamic graphical status summaries for nucelotide sequencing
US20230215515A1 (en) 2021-12-23 2023-07-06 Illumina Software, Inc. Facilitating secure execution of external workflows for genomic sequencing diagnostics
WO2023129764A1 (en) 2021-12-29 2023-07-06 Illumina Software, Inc. Automatically switching variant analysis model versions for genomic analysis applications

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5572598A (en) * 1991-08-22 1996-11-05 Kla Instruments Corporation Automated photomask inspection apparatus
US5631734A (en) * 1994-02-10 1997-05-20 Affymetrix, Inc. Method and apparatus for detection of fluorescently labeled materials

Family Cites Families (93)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3216313A (en) * 1961-06-23 1965-11-09 Bausch & Lomb Monochromator of the type having a plane grating therein
GB1077501A (en) * 1964-02-04 1967-08-02 Nat Res Dev Scanning spectrophotometer
US3802966A (en) * 1969-08-22 1974-04-09 Ethyl Corp Apparatus for delivering a fluid suspension to a forming unit clear reactor power plant
US3632212A (en) * 1970-06-01 1972-01-04 Honeywell Inc Gas temperature measurement system employing a laser
AT353497B (en) * 1972-05-23 1979-11-12 Leitz Ernst Gmbh DEVICE ON MICROSCOPES FOR AUTOMATIC FOCUSING OF THE DEVICE ON DIFFERENT OBJECT LEVELS
US3984171A (en) * 1974-08-21 1976-10-05 Image Information Inc. Linear scan system
JPS6112373B2 (en) * 1974-09-04 1986-04-08 Hitachi Ltd
US4070111A (en) * 1976-06-10 1978-01-24 Nicolas James Harrick Rapid scan spectrophotometer
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
US4204929A (en) * 1978-04-18 1980-05-27 University Patents, Inc. Isoelectric focusing method
US4176925A (en) * 1978-06-07 1979-12-04 Gte Laboratories Incorporated Laser scanner for photolithography of slotted mask color cathode ray tubes
US4342905A (en) * 1979-08-31 1982-08-03 Nippon Kogaku K.K. Automatic focusing device of a microscope
JPS5817767A (en) * 1981-07-23 1983-02-02 Fuji Photo Film Co Ltd Picture scanner
AU1944283A (en) * 1982-08-27 1984-03-29 Ekins Roger Philip Measurement of analyte concentration
DE3245939C2 (en) * 1982-12-11 1985-12-19 Fa. Carl Zeiss, 7920 Heidenheim Device for generating an image of the fundus
US4537861A (en) * 1983-02-03 1985-08-27 Elings Virgil B Apparatus and method for homogeneous immunoassay
US4626684A (en) * 1983-07-13 1986-12-02 Landa Isaac J Rapid and automatic fluorescence immunoassay analyzer for multiple micro-samples
US4580895A (en) * 1983-10-28 1986-04-08 Dynatech Laboratories, Incorporated Sample-scanning photometer
DE3422143A1 (en) * 1984-06-14 1985-12-19 Josef Prof. Dr. Bille WAFER INSPECTION DEVICE
GB8429212D0 (en) * 1984-11-19 1984-12-27 Vincent Patents Ltd Exhaust systems for ic engines
EP0194132A3 (en) * 1985-03-06 1988-08-03 Murex Corporation Imaging immunoassay detection system and method
JPS61290311A (en) * 1985-06-19 1986-12-20 Hitachi Ltd Apparatus and method for inspecting soldered zone
DD254998A1 (en) * 1985-07-26 1988-03-16 Zeiss Jena Veb Carl ARRANGEMENT FOR THE IMAGE AND ANALYSIS OF FLUORESCENCE SIGNALS
US4963498A (en) * 1985-08-05 1990-10-16 Biotrack Capillary flow device
US5164598A (en) * 1985-08-05 1992-11-17 Biotrack Capillary flow device
GB8619206D0 (en) * 1986-08-06 1986-09-17 Ekins Roger Philip Fluorometric determination of analyte concentration
JPS63167313A (en) * 1986-12-27 1988-07-11 Hitachi Ltd Automatic focus control method
JPH0750094B2 (en) * 1987-01-28 1995-05-31 富士写真フイルム株式会社 Continuous manufacturing method for chemical analysis slides
DE3724878A1 (en) 1987-07-28 1989-02-09 Manfred Klein LIFT VACUUM CLEANER FOR A TRANSPORTATION DEVICE
GB8803000D0 (en) * 1988-02-10 1988-03-09 Ekins Roger Philip Determination of ambient concentrations of several analytes
US4844617A (en) * 1988-01-20 1989-07-04 Tencor Instruments Confocal measuring microscope with automatic focusing
US5188963A (en) * 1989-11-17 1993-02-23 Gene Tec Corporation Device for processing biological specimens for analysis of nucleic acids
US5320808A (en) * 1988-08-02 1994-06-14 Abbott Laboratories Reaction cartridge and carousel for biological sample analyzer
US5281516A (en) * 1988-08-02 1994-01-25 Gene Tec Corporation Temperature control apparatus and method
US5382511A (en) * 1988-08-02 1995-01-17 Gene Tec Corporation Method for studying nucleic acids within immobilized specimens
US5200051A (en) * 1988-11-14 1993-04-06 I-Stat Corporation Wholly microfabricated biosensors and process for the manufacture and use thereof
JPH036444A (en) * 1989-06-02 1991-01-11 Furoobell:Kk Inspecting apparatus by fluorescence
US5424186A (en) * 1989-06-07 1995-06-13 Affymax Technologies N.V. Very large scale immobilized polymer synthesis
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
US5061075A (en) * 1989-08-07 1991-10-29 Alfano Robert R Optical method and apparatus for diagnosing human spermatozoa
US5034613A (en) * 1989-11-14 1991-07-23 Cornell Research Foundation, Inc. Two-photon laser microscopy
US5346672A (en) * 1989-11-17 1994-09-13 Gene Tec Corporation Devices for containing biological specimens for thermal processing
US5091652A (en) * 1990-01-12 1992-02-25 The Regents Of The University Of California Laser excited confocal microscope fluorescence scanner and method
US5132524A (en) * 1990-05-21 1992-07-21 Lazerdata Corporation Multi directional laser scanner
JP3001612B2 (en) * 1990-06-22 2000-01-24 ファナック株式会社 Operation control device of scanner galvanometer
GB9014263D0 (en) * 1990-06-27 1990-08-15 Dixon Arthur E Apparatus and method for spatially- and spectrally- resolvedmeasurements
GB9015793D0 (en) * 1990-07-18 1990-09-05 Medical Res Council Confocal scanning optical microscope
CA2097708A1 (en) * 1990-12-06 1992-06-07 Stephen P. A. Fodor Very large scale immobilized polymer synthesis
US5198871A (en) * 1991-06-18 1993-03-30 Southwest Research Institute Laser-induced-fluorescence inspection of jet fuels
JPH0511192A (en) * 1991-07-08 1993-01-19 Nikon Corp Laser scanning microscope
AU2422492A (en) * 1991-08-07 1993-03-02 H & N Instruments, Inc. Synthesis of chain chemical compounds
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
US5384261A (en) * 1991-11-22 1995-01-24 Affymax Technologies N.V. Very large scale immobilized polymer synthesis using mechanically directed flow paths
WO1993009668A1 (en) * 1991-11-22 1993-05-27 Affymax Technology N.V. Combinatorial strategies for polymer synthesis
JP2511391B2 (en) * 1991-12-04 1996-06-26 シーメンス アクチエンゲゼルシヤフト Optical distance sensor
US5310469A (en) * 1991-12-31 1994-05-10 Abbott Laboratories Biosensor with a membrane containing biologically active material
GB9218482D0 (en) 1992-09-01 1992-10-14 Dixon Arthur E Apparatus and method for scanning laser imaging of macroscopic samples
US5235180A (en) * 1992-03-05 1993-08-10 General Scanning, Inc. Rotary motor having an angular position transducer and galvanometer scanning system employing such motor
US5296703A (en) * 1992-04-01 1994-03-22 The Regents Of The University Of California Scanning confocal microscope using fluorescence detection
JP3616107B2 (en) * 1992-04-10 2005-02-02 株式会社日立製作所 Polynucleotide detection method
US5248876A (en) * 1992-04-21 1993-09-28 International Business Machines Corporation Tandem linear scanning confocal imaging system with focal volumes at different heights
US5304487A (en) * 1992-05-01 1994-04-19 Trustees Of The University Of Pennsylvania Fluid handling in mesoscale analytical devices
US5498392A (en) * 1992-05-01 1996-03-12 Trustees Of The University Of Pennsylvania Mesoscale polynucleotide amplification device and method
US5486335A (en) * 1992-05-01 1996-01-23 Trustees Of The University Of Pennsylvania Analysis based on flow restriction
JPH06167443A (en) * 1992-10-23 1994-06-14 Olympus Optical Co Ltd Measuring apparatus utilizing surface plasmon resonance
JPH06148525A (en) * 1992-10-30 1994-05-27 Koike Seiki Kk Confocal laser microscope
JP3343276B2 (en) * 1993-04-15 2002-11-11 興和株式会社 Laser scanning optical microscope
US5424841A (en) * 1993-05-28 1995-06-13 Molecular Dynamics Apparatus for measuring spatial distribution of fluorescence on a substrate
DE69418248T2 (en) * 1993-06-03 1999-10-14 Hamamatsu Photonics Kk Optical laser scanning system with Axikon
US5479252A (en) 1993-06-17 1995-12-26 Ultrapointe Corporation Laser imaging system for inspection and analysis of sub-micron particles
US5483055A (en) 1994-01-18 1996-01-09 Thompson; Timothy V. Method and apparatus for performing an automatic focus operation for a microscope
JP3176770B2 (en) * 1993-06-28 2001-06-18 キヤノン株式会社 Fluid inspection device
US5381224A (en) 1993-08-30 1995-01-10 A. E. Dixon Scanning laser imaging system
JPH0763508A (en) * 1993-08-31 1995-03-10 Ishikawajima Harima Heavy Ind Co Ltd Laser microscope
US5532873A (en) 1993-09-08 1996-07-02 Dixon; Arthur E. Scanning beam laser microscope with wide range of magnification
US5494124A (en) * 1993-10-08 1996-02-27 Vortexx Group, Inc. Negative pressure vortex nozzle
AU8126694A (en) * 1993-10-26 1995-05-22 Affymax Technologies N.V. Arrays of nucleic acid probes on biological chips
JPH07209187A (en) * 1993-11-30 1995-08-11 Omron Corp Laser scanning type cell analyser
US5578832A (en) * 1994-09-02 1996-11-26 Affymetrix, Inc. Method and apparatus for imaging a sample on a device
JPH07261097A (en) * 1994-03-17 1995-10-13 Fujitsu Ltd Microscope and image acquiring method
JP3398464B2 (en) * 1994-03-17 2003-04-21 ソニー株式会社 Laser microscope
US5571639A (en) * 1994-05-24 1996-11-05 Affymax Technologies N.V. Computer-aided engineering system for design of sequence arrays and lithographic masks
JP3568626B2 (en) * 1994-05-24 2004-09-22 オリンパス株式会社 Scanning optical microscope
EP0695941B1 (en) 1994-06-08 2002-07-31 Affymetrix, Inc. Method and apparatus for packaging a chip
US5866911A (en) * 1994-07-15 1999-02-02 Baer; Stephen C. Method and apparatus for improving resolution in scanned optical system
US5459325A (en) * 1994-07-19 1995-10-17 Molecular Dynamics, Inc. High-speed fluorescence scanner
JPH0862123A (en) * 1994-08-23 1996-03-08 Olympus Optical Co Ltd Scanning optical measuring apparatus
WO1996018205A1 (en) * 1994-12-08 1996-06-13 Molecular Dynamics, Inc. Fluorescence imaging system employing a macro scanning objective
US5585639A (en) * 1995-07-27 1996-12-17 Hewlett-Packard Company Optical scanning apparatus
US5646411A (en) * 1996-02-01 1997-07-08 Molecular Dynamics, Inc. Fluorescence imaging system compatible with macro and micro scanning objectives
WO1997043611A1 (en) * 1996-05-16 1997-11-20 Affymetrix, Inc. Systems and methods for detection of labeled materials

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5572598A (en) * 1991-08-22 1996-11-05 Kla Instruments Corporation Automated photomask inspection apparatus
US5631734A (en) * 1994-02-10 1997-05-20 Affymetrix, Inc. Method and apparatus for detection of fluorescently labeled materials

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP0902885A4 *

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
USRE43097E1 (en) 1994-10-13 2012-01-10 Illumina, Inc. Massively parallel signature sequencing by ligation of encoded adaptors
US9273354B2 (en) 1997-05-23 2016-03-01 Illumina, Inc. System and apparatus for sequential processing of analytes
WO1999052625A3 (en) * 1998-04-09 2000-01-06 Diagnostikforschung Inst Method and device for producing synthesis or analysis series
WO1999052625A2 (en) * 1998-04-09 1999-10-21 Institut für Diagnostikforschung GmbH an der Freien Universität Berlin Method and device for producing synthesis or analysis series
WO1999063328A1 (en) * 1998-06-01 1999-12-09 Abbott Laboratories Sensor utilizing raman spectroscopy for non-invasive monitoring of analytes in biological fluid and method of use
US6064897A (en) * 1998-06-01 2000-05-16 Abbott Laboratories Sensor utilizing Raman spectroscopy for non-invasive monitoring of analytes in biological fluid and method of use
WO2000063678A1 (en) * 1999-04-14 2000-10-26 Carl Zeiss Jena Gmbh Device for examining samples
DE19916749B4 (en) * 1999-04-14 2004-02-12 Carl Zeiss Jena Gmbh Procedure for examining samples
US6823079B1 (en) 1999-04-14 2004-11-23 Carl Zeiss Jena Gmbh Device for examining samples
JP2001091463A (en) * 1999-09-22 2001-04-06 Tosoh Corp Scanner type fluorescence detector using small-sized exciting light source
US7033840B1 (en) 1999-11-09 2006-04-25 Sri International Reaction calorimeter and differential scanning calorimeter for the high-throughput synthesis, screening and characterization of combinatorial libraries
US6602714B1 (en) 1999-11-09 2003-08-05 Sri International Viscosity and mass sensor for the high-throughput synthesis, screening and characterization of combinatorial libraries
EP1311824A1 (en) * 2000-06-25 2003-05-21 Affymetrix, Inc. Optically active substrates
EP1311824A4 (en) * 2000-06-25 2010-12-08 Affymetrix Inc Optically active substrates
US6750963B2 (en) 2002-05-21 2004-06-15 Agilent Technologies, Inc. Imaging systems for signals on a surface
DE10236433A1 (en) * 2002-08-06 2004-02-19 Förderverein Institut für Medizintechnik Dresden e.V. Device for optically detecting liquid samples in droplet form formed discretely on planar substrate or in cavities of substrate comprises optical detector arranged in optical axis of light beam, and optical element
EP2291539A1 (en) * 2008-06-25 2011-03-09 Real-Time Genomics, Llc Method and apparatus for melting curve analysis of nucleic acids in microarray format
EP2291539A4 (en) * 2008-06-25 2013-05-01 Real Time Genomics Llc Method and apparatus for melting curve analysis of nucleic acids in microarray format
US9103785B2 (en) 2008-06-25 2015-08-11 Emergence Genomics, Llc Method and apparatus for melting curve analysis of nucleic acids in microarray format
CN104568873A (en) * 2014-12-22 2015-04-29 中国科学院苏州生物医学工程技术研究所 Laser scanning confocal microscope for imaging fluorescent substances
CN108885260A (en) * 2016-04-08 2018-11-23 苹果公司 Transition time detector with single shaft scanning
CN108885260B (en) * 2016-04-08 2022-06-03 苹果公司 Time-of-flight detector with single axis scanning
WO2018033523A1 (en) * 2016-08-16 2018-02-22 Technische Universität Hamburg-Harburg Application profile reactor for operando measurements
US10746661B2 (en) 2016-08-16 2020-08-18 REACNOSTICS GmbH Profile reactor for operando measurements

Also Published As

Publication number Publication date
US20010030290A1 (en) 2001-10-18
EP0902885A1 (en) 1999-03-24
JP2006258821A (en) 2006-09-28
US5981956A (en) 1999-11-09
JP2000512744A (en) 2000-09-26
US20020185610A1 (en) 2002-12-12
US6207960B1 (en) 2001-03-27
EP0902885A4 (en) 2006-09-27
US6597000B2 (en) 2003-07-22

Similar Documents

Publication Publication Date Title
US5981956A (en) Systems and methods for detection of labeled materials
US5631734A (en) Method and apparatus for detection of fluorescently labeled materials
US7158224B2 (en) Optically active substrates
US6741344B1 (en) Method and apparatus for detection of fluorescently labeled materials
US5578832A (en) Method and apparatus for imaging a sample on a device
US6704104B2 (en) Multi-wavelength array reader for biological assay
KR100339379B1 (en) biochip and apparatus and method for measuring biomaterial of the same
US7154598B2 (en) Excitation and imaging of fluorescent arrays
US5812272A (en) Apparatus and method with tiled light source array for integrated assay sensing
US20040048362A1 (en) Method and apparatus for imaging a sample on a device
US20050123907A1 (en) Methods for making a device for concurrently processing multiple biological chip assays
JP2009515192A (en) Optical fiber exploration microslide, microslide kit, and use thereof
US20020074512A1 (en) High performance substrate scanning
JPH11505015A (en) Multi-capillary fluorescence detection system
CN1311436A (en) Reading of biological chip fluorescent image on rotary platform
US6587197B1 (en) Multiple microchannels chip for biomolecule imaging, and method of use thereof
JP2002508857A (en) Specimen irradiation apparatus using optical cavity for dark field irradiation and method of use
US20030232427A1 (en) Optically active substrates for examination of biological materials
US6670198B2 (en) Test piece and system for reading out image information from the test piece
JP2002148265A (en) Biochemical analysis method, unit for biochemical analysis used therefor, and target detector for detecting target from unit for the biochemical analysis

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): JP

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FI FR GB GR IE IT LU MC NL PT SE

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 1997925604

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1997925604

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1997925604

Country of ref document: EP