US20150298089A1 - Systems for Filling a Sample Array by Droplet Dragging - Google Patents

Systems for Filling a Sample Array by Droplet Dragging Download PDF

Info

Publication number
US20150298089A1
US20150298089A1 US14/582,122 US201414582122A US2015298089A1 US 20150298089 A1 US20150298089 A1 US 20150298089A1 US 201414582122 A US201414582122 A US 201414582122A US 2015298089 A1 US2015298089 A1 US 2015298089A1
Authority
US
United States
Prior art keywords
array
holes
hole
liquid
arrays
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US14/582,122
Other versions
US20170028376A9 (en
Inventor
Ian Hunter
Colin J.H. Brenan
Tanya S. Kanigan
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Massachusetts Institute of Technology
Original Assignee
Massachusetts Institute of Technology
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US09/850,123 external-priority patent/US6893877B2/en
Application filed by Massachusetts Institute of Technology filed Critical Massachusetts Institute of Technology
Priority to US14/582,122 priority Critical patent/US20170028376A9/en
Publication of US20150298089A1 publication Critical patent/US20150298089A1/en
Publication of US20170028376A9 publication Critical patent/US20170028376A9/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J19/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J19/0046Sequential or parallel reactions, e.g. for the synthesis of polypeptides or polynucleotides; Apparatus and devices for combinatorial chemistry or for making molecular arrays
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01FMIXING, e.g. DISSOLVING, EMULSIFYING OR DISPERSING
    • B01F33/00Other mixers; Mixing plants; Combinations of mixers
    • B01F33/30Micromixers
    • B01F33/302Micromixers the materials to be mixed flowing in the form of droplets
    • B01F33/3021Micromixers the materials to be mixed flowing in the form of droplets the components to be mixed being combined in a single independent droplet, e.g. these droplets being divided by a non-miscible fluid or consisting of independent droplets
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01FMIXING, e.g. DISSOLVING, EMULSIFYING OR DISPERSING
    • B01F33/00Other mixers; Mixing plants; Combinations of mixers
    • B01F33/30Micromixers
    • B01F33/3035Micromixers using surface tension to mix, move or hold the fluids
    • B01F33/30351Micromixers using surface tension to mix, move or hold the fluids using hydrophilic/hydrophobic surfaces
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L3/00Containers or dishes for laboratory use, e.g. laboratory glassware; Droppers
    • B01L3/50Containers for the purpose of retaining a material to be analysed, e.g. test tubes
    • B01L3/502Containers for the purpose of retaining a material to be analysed, e.g. test tubes with fluid transport, e.g. in multi-compartment structures
    • B01L3/5025Containers for the purpose of retaining a material to be analysed, e.g. test tubes with fluid transport, e.g. in multi-compartment structures for parallel transport of multiple samples
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L3/00Containers or dishes for laboratory use, e.g. laboratory glassware; Droppers
    • B01L3/50Containers for the purpose of retaining a material to be analysed, e.g. test tubes
    • B01L3/508Containers for the purpose of retaining a material to be analysed, e.g. test tubes rigid containers not provided for above
    • B01L3/5085Containers for the purpose of retaining a material to be analysed, e.g. test tubes rigid containers not provided for above for multiple samples, e.g. microtitration plates
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L3/00Containers or dishes for laboratory use, e.g. laboratory glassware; Droppers
    • B01L3/50Containers for the purpose of retaining a material to be analysed, e.g. test tubes
    • B01L3/508Containers for the purpose of retaining a material to be analysed, e.g. test tubes rigid containers not provided for above
    • B01L3/5085Containers for the purpose of retaining a material to be analysed, e.g. test tubes rigid containers not provided for above for multiple samples, e.g. microtitration plates
    • B01L3/50857Containers for the purpose of retaining a material to be analysed, e.g. test tubes rigid containers not provided for above for multiple samples, e.g. microtitration plates using arrays or bundles of open capillaries for holding samples
    • 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
    • C12Q1/34Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving hydrolase
    • 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
    • G01N21/75Systems in which material is subjected to a chemical reaction, the progress or the result of the reaction being investigated
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N35/00Automatic analysis not limited to methods or materials provided for in any single one of groups G01N1/00 - G01N33/00; Handling materials therefor
    • G01N35/10Devices for transferring samples or any liquids to, in, or from, the analysis apparatus, e.g. suction devices, injection devices
    • G01N35/1002Reagent dispensers
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00277Apparatus
    • B01J2219/00279Features relating to reactor vessels
    • B01J2219/00306Reactor vessels in a multiple arrangement
    • B01J2219/00313Reactor vessels in a multiple arrangement the reactor vessels being formed by arrays of wells in blocks
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00277Apparatus
    • B01J2219/00279Features relating to reactor vessels
    • B01J2219/00306Reactor vessels in a multiple arrangement
    • B01J2219/00313Reactor vessels in a multiple arrangement the reactor vessels being formed by arrays of wells in blocks
    • B01J2219/00315Microtiter plates
    • B01J2219/00317Microwell devices, i.e. having large numbers of wells
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00277Apparatus
    • B01J2219/00351Means for dispensing and evacuation of reagents
    • 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
    • B01J2219/00533Sheets essentially rectangular
    • 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
    • B01J2219/00536Sheets in the shape of disks
    • 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
    • B01J2219/0054Means for coding or tagging the apparatus or the reagents
    • B01J2219/00547Bar codes
    • B01J2219/005492-dimensional
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00585Parallel processes
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00599Solution-phase processes
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L2200/00Solutions for specific problems relating to chemical or physical laboratory apparatus
    • B01L2200/06Fluid handling related problems
    • B01L2200/0642Filling fluids into wells by specific techniques
    • CCHEMISTRY; METALLURGY
    • C40COMBINATORIAL TECHNOLOGY
    • C40BCOMBINATORIAL CHEMISTRY; LIBRARIES, e.g. CHEMICAL LIBRARIES
    • C40B60/00Apparatus specially adapted for use in combinatorial chemistry or with libraries
    • C40B60/14Apparatus specially adapted for use in combinatorial chemistry or with libraries for creating libraries
    • CCHEMISTRY; METALLURGY
    • C40COMBINATORIAL TECHNOLOGY
    • C40BCOMBINATORIAL CHEMISTRY; LIBRARIES, e.g. CHEMICAL LIBRARIES
    • C40B70/00Tags or labels specially adapted for combinatorial chemistry or libraries, e.g. fluorescent tags or bar codes
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N35/00Automatic analysis not limited to methods or materials provided for in any single one of groups G01N1/00 - G01N33/00; Handling materials therefor
    • G01N2035/00178Special arrangements of analysers
    • G01N2035/00237Handling microquantities of analyte, e.g. microvalves, capillary networks
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N35/00Automatic analysis not limited to methods or materials provided for in any single one of groups G01N1/00 - G01N33/00; Handling materials therefor
    • G01N35/00029Automatic analysis not limited to methods or materials provided for in any single one of groups G01N1/00 - G01N33/00; Handling materials therefor provided with flat sample substrates, e.g. slides
    • G01N35/00069Automatic analysis not limited to methods or materials provided for in any single one of groups G01N1/00 - G01N33/00; Handling materials therefor provided with flat sample substrates, e.g. slides whereby the sample substrate is of the bio-disk type, i.e. having the format of an optical disk
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10TTECHNICAL SUBJECTS COVERED BY FORMER US CLASSIFICATION
    • Y10T436/00Chemistry: analytical and immunological testing
    • Y10T436/11Automated chemical analysis
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10TTECHNICAL SUBJECTS COVERED BY FORMER US CLASSIFICATION
    • Y10T436/00Chemistry: analytical and immunological testing
    • Y10T436/11Automated chemical analysis
    • Y10T436/112499Automated chemical analysis with sample on test slide
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10TTECHNICAL SUBJECTS COVERED BY FORMER US CLASSIFICATION
    • Y10T436/00Chemistry: analytical and immunological testing
    • Y10T436/25Chemistry: analytical and immunological testing including sample preparation

Definitions

  • the present invention pertains to methods for manufacturing and using apparatus for manipulating, transporting, and analyzing a large number of microscopic samples of a liquid or of materials including cells currently or formerly in liquid suspension.
  • a method and an array filling system are provided for loading a plurality of disparate sample containers, the sample containers comprising an integral structure.
  • each receptacle is characterized by a hydrophilic surface, and the receptacles are separated by a hydrophobic surface.
  • the system has a liquid transfer device capable of holding liquid and adapted for motion to cause sequential communication of liquid held in the liquid transfer device with successive receptacles of the array by dragging the liquid across the hydrophobic surface.
  • the array filling system has an array of liquid transfer devices capable of holding liquid, the plurality of liquid transfer devices adapted for motion to cause sequential communication of liquid held in the liquid transfer devices with successive receptacles of the array by dragging the liquid across the hydrophobic surface.
  • a method for transferring a liquid into an array of receptacles having hydrophilic surfaces and separated by a hydrophobic surface has steps of:
  • the method may have a further step of withdrawing residual fluid from the surface of the platen.
  • the step of causing communication of liquid in the liquid transfer device with the hydrophilic surfaces of the plurality of hydrophilic receptacles may include forming a droplet of the liquid on the end of a capillary, or on the end of a pipette or needle, and may, further, include transferring liquid into through-holes of an array of through-holes.
  • FIGS. 1 a and 1 b show a top and exploded cross-sectional views, respectively, of a high-density array of through-holes in accordance with one embodiment of the present invention
  • FIG. 1 c shows a schematic side view of a continuous sheet array of through-holes in accordance with another embodiment of the present invention
  • FIG. 2 a is top view of a portion of the platen of FIG. 1 a in which the through-holes are configured on rectangular centers;
  • FIG. 2 b is top view of a portion of the platen of FIG. 1 a in which the through-holes are configured in a hexagonal close-packed array;
  • FIG. 3 is a further side view in cross-section of a portion of a laminated platen containing multiple through-holes for analysis of liquid samples in accordance with a preferred embodiment of the present invention
  • FIG. 4 is a top view of round sample wafer populated with through-holes in accordance with an embodiment of the present invention.
  • FIGS. 5 a - 5 d show examples of arrays with through-hole volumes that are a function of array position
  • FIGS. 5 e and 5 f show cross sections of through-hole prisms as employed for massively parallel liquid chromatography or electrophoresis, in accordance with embodiments of the present invention
  • FIG. 6 shows an example of an interlocking array
  • FIG. 7 depicts the configuration of a compression mold for compression molding on an array of through-holes in accordance with an embodiment of the present invention
  • FIG. 8 is a flow chart depicting steps in the fabrication of a through-hole array by EDM, in accordance with an embodiment of the invention.
  • FIGS. 9 a - 9 c depict a sequence of operations for filling a through-hole array with a first liquid
  • FIG. 10 is a schematic depiction of a method for sampling the contents of a large-format microtiter plate and loading a subarray of the through-hole array in accordance with an embodiment of the present invention
  • FIG. 11 is a schematic depiction of a method for sampling the contents of a large-format microtiter plate and loading subarrays of multiple through-hole arrays in accordance with an embodiment of the present invention
  • FIG. 12 a depicts parallel loading of subarrays of multiple stacked microwell arrays in accordance with embodiments of the present invention
  • FIGS. 12 b and 12 c depict parallel loading of subarrays of a microwell array by means of flexible members, in accordance with embodiments of the present invention
  • FIG. 13 is a flow chart depicting a method for successive dilution of the contents of a microwell array in accordance with an embodiment of the present invention
  • FIGS. 14 a - 14 c depict a sequence of operations for exposing respective through-holes of a through-hole array to a second liquid and for creating a specified gradient of a specified characteristic with respect to placement of the through-holes in the array in accordance with an embodiment of the present invention
  • FIG. 14 d shows a side view is shown of a through-hole array plate with a graduated filter for providing a concentration gradient in accordance with an embodiment of the invention
  • FIG. 15 depicts mixing of the contents of a through-hole array with the contents of another through-hole array brought into registration of the through-holes in accordance with embodiments of the present invention
  • FIGS. 16 and 17 depict further stages in the mixing of the contents of a through-hole array with the contents of another through-hole array brought into registration of the through-holes in accordance with embodiments of the present invention
  • FIG. 18 depicts mixing of the contents of a through-hole array with the contents of another through-hole array by application of external pressure, in accordance with embodiments of the present invention
  • FIG. 19 depicts mixing of the contents of a fully filled through-hole array with the contents of another incompletely filled through-hole array brought into registration of the through-holes in accordance with embodiments of the present invention
  • FIG. 20 shows plots of calculated times for evaporation of a water droplet as a function of the ambient relative humidity
  • FIG. 21 depicts a humidity chamber for loading and unloading through-holes while maintaining a high relative humidity in the environment surrounding the array, in accordance with an embodiment of the present invention
  • FIG. 22 a depicts an exploded perspective view of a portable humidity chamber for preventing evaporation of fluid from the arrays during fluorescent imaging analysis, incubation and transferring the array between other humidified environments;
  • FIG. 22 b is a cross-sectional side view of the a portable humidity chamber of FIG. 22 a.
  • methods are provided for producing different chemical reactions within an array of through-holes.
  • the invention is advantageously employed, for example, in screening operations, where different reaction conditions are advantageously provided among the various through-holes of the array.
  • different chemical species may be loaded into different through-holes of the array, and concentrations of the various species might also be differentiated among the various through-holes.
  • the invention may thus provide a method for screening compound libraries, for example, to predict the ability of each compound to be absorbed by a patient.
  • FIG. 1 a shows a top view of a platen 10 , otherwise referred to herein as a “substrate,” “sample wafer,” or “through-hole plate.”
  • Platen 10 is the carrier of a large number of through-holes 12 which traverse platen 10 from a top surface 14 to an opposing surface 16 of the platen, as shown in the cross-sectional side view of FIG. 1 b .
  • platen may refer to a structure having substantially parallel plane surfaces and transverse dimensions substantially exceeding the thickness of the structure, alternative geometries are within the scope of the present invention, and use of the term “platen” is not restrictive.
  • a prism-shaped geometry is described below, for example.
  • Through-holes 12 constitute assay wells (or “microwells”) in accordance with an embodiment of the invention.
  • Through-holes 12 may be shaped as circular right cylinders, or, alternatively, may have rectangular cross-sections. Otherwise-shaped through-holes are also within the scope of the present invention.
  • through-hole plate 10 is preferably made of conductive silicon, other types of rigid materials, such as metal, glass, or plastic may be used provided that the material is chemically inert with respect to the sample substances, or can be rendered so by appropriate surface treatments.
  • Each through-hole 12 is typically of a substantially square cross-section, although geometries, such as circular or rectangular cross-sections may be used. Through-holes 12 are also referred to herein as “channels.”
  • Through-holes 12 may be centered on a rectangular grid, as shown in FIG. 2 a , or in a close-packed hexagonal lattice, as shown in FIG. 2 b .
  • a typical thickness 20 of platen 10 is on the order of 0.5-2 mm, while through-holes 12 have typical characteristic dimensions (such as diameters) 36 of on the order of 100-400 ⁇ m.
  • the volume of each through-hole 12 between surface 14 and surface 16 is on the order of ⁇ 10 ⁇ 7 cm 3 or greater.
  • Through-holes 12 are spaced on centers typically on the order of twice the diameter of the holes, although all spacing configurations are within the scope of the invention and of the appended claims.
  • a hole-to-hole spacing 38 of 500 ⁇ m is typically employed, which corresponds to an array density of 400 holes per square centimeter of plate.
  • microwells are produced for the assay of a chemical or biochemical reaction where the volume of each microwell may be less than 100 nanoliters (10 ⁇ 7 cm 3 ). The packing density of wells may thereby be increased by several orders of magnitude over prior art technology.
  • Grouping of through-holes into smaller sub-arrays may also be used, and, more particularly, a reproducible pattern may be applied to a plurality of the sub-arrays.
  • Each through-hole 12 may be identified by its own address within the array.
  • platen 10 may also advantageously be formed of a laminate of materials, with a central layer 24 and outer “sandwiching” layers 22 .
  • exterior sections 22 of the microwell, adjacent to surfaces 14 and 16 of platen 10 have a hydrophobic wall surface in accordance with a preferred embodiment of the invention, while the interior section 24 of the through-hole wall has a hydrophilic surface thereby preferentially attracting an aqueous liquid sample.
  • an array having hydrophilic faces and hydrophobic through-holes may be uniformly filled with a low surface tension liquid such as an alkane.
  • each well is typically overfilled by about 10% above the volume surrounded by the four walls of the microwell. Under these circumstances, liquid sample 30 may form convex meniscus surfaces 34 on both the upper and lower surfaces of the sample.
  • An underfilled microwell 26 will typically be characterized by a liquid sample exhibiting a concave meniscus 28 on both the upper and lower surfaces of liquid sample 30 .
  • the apertures of through-holes 12 need not be square, and, in accordance with an alternate embodiment of the present invention, flanges 8 may extend above planar surface 14 surrounding some or all of through-holes 12 while indentations 6 may be fabricated rounding the edges of through-holes 12 at opposing surface 16 .
  • Flanges 8 and indentations 6 may advantageously provide for registration of successive platens 10 , in the case where platens are stacked, and in processes of mixing or dilution, as discussed in detail below.
  • Through-holes 12 may be loaded with a first sample 18 in liquid form.
  • Sample 18 is allowed to react with a second sample where the second sample may include a variety of test samples and by subsequent or concurrent analysis of the reaction products, using, for example, optical markers, a large number of reactions may be processed and analyzed in parallel.
  • first sample 18 may be a solution containing pharmacologically relevant proteins or other molecules.
  • a solution may include, for example, cells in aqueous suspension, eukaryotic (animal, yeast) or prokaryotic (bacteria) cells, hybrid cells, and biological molecules including, for example, antibodies and enzymes, although application to other biological or non-biological assays is within the scope of the invention as claimed herein. All such reagents may also be referred to herein and in the appended claims as “targets.”
  • targets Typical yeast cell concentrations of 10 7 cells per milliliter of solution yield on the order of 1000 cells per 100 nanoliter well.
  • an entire chip or the subset of through-hole wells constituting a contiguous region of platen 10 may be populated with a single strain of cells.
  • a typical procedure assay procedure such as may be employed in pharmaceutical research, entails the subsequent addressed introduction of a test sample including one or more analytes into the through-hole wells, with selected materials introduced into subsets of through-holes that may include one or more through-holes.
  • the test sample addressably introduced into the subsets of through-holes may contain drug candidates or known drugs.
  • the test sample may be comprised of multiple components, introduced at the same time or sequentially. Components of the test sample may include analytes, antagonists, reagents, solvents, or any other materials and may be introduced in liquid form or otherwise.
  • test samples are introduced into the through-hole wells in liquid form in order to facilitate rapid reaction via diffusion with first sample 18 already resident in liquid form in the through-holes.
  • libraries The set of substances from which the second sample addressed to a particular through-hole site is drawn is referred to in this description and in the appended claims as a “library” of substances.
  • the library is of a substantial size and thus advantageously utilizes the capability of the present invention to facilitate parallel reaction and analysis of large numbers of substances.
  • libraries may be composed of between 10 3 and 10 9 substances and combinations of substances.
  • the throughput of a screening system incorporating arrays of through-hole sample containers may be further increased by using a continuous sheet 300 of through-holes 12 .
  • the interiors of the through-holes may be hydrophilic, as described above in reference to FIG. 1 b , and the surface of sheet 300 may be hydrophobic, as also described.
  • Through-holes 12 may be filled in a continuous manner by passing sheet 300 through an aqueous medium 302 contained in fluid trough 304 . After through-holes 12 have been filled, sheet 300 may be wound onto a spool 306 or cassette for storage followed by assay, or, alternatively, may be assayed directly.
  • Assays may be by optical readout such as fluorescence absorbance or chemi-luminescence measurements, all of which may be performed by passing the sheet across an optical detector such as a CCD array.
  • Sample array sheets 300 are preferably produced with registered holes, either by precision production processes or in matched sets.
  • the sheet composition may be a polymer, elastomer, or metal, including an amorphous metal. Multiple sheets may be mated in the same way that platens of through-holes may be stacked, as described below, for example, with reference to FIGS. 16-19 , in order to initiate reactions, or for other purposes.
  • FIG. 1 c shows a second sheet 308 of through-holes 310 being brought into contact with sheet 300 in mixing area 312 for mixing of the liquid contents of the respective through-holes. Examples of applications involving this embodiment include, screening genetic libraries or screening combinatorial chemical libraries contained on polymer beads.
  • inventions of the invention may advantageously include extremely high throughput, reduction or elimination of high-cost automation components, and the small size of a screening system with sample handling and detection modules.
  • the holes in the sheet may be, if desired, produced online with an array of punches or UV lasers.
  • an E. coli genetic library is prepared containing mutations in the beta galactosidase enzyme.
  • the E. coli cells are grown to a density in phosphate-limited media such that there is an average of 1 cell for every 200 nl of liquid.
  • the media also contains, MUG, a fluorogenic substrate for beta-galactosidase.
  • a through-hole sheet is prepared with a hydrophobic exterior and hydrophilic through-holes at a density of 10 ⁇ 7 per square meter. Registration holes are includes in the tape to aid in precise dispensing. Each through-hole holds 70 nl of fluid.
  • a spool of the through-hole sheet is unwound and guided through a trough containing the cell solution, so that each through hole is filled.
  • the sheet is then wound onto spacers and into a receiving spool.
  • the spacers prevent smearing of the liquid and provide for gas transfer in and out of the spool.
  • the spool is incubated in a humidified enviroment at 37° C. for 24 hours.
  • the spool is then unwound as it is passed between a uniform photo-illumination source with a wavelength of 350 nm and a CCD imaging system with a 450 nm filter.
  • the position in the sheet of colonies with exceptionally high enzyme activity is recorded and those colonies are retrieved using a robotic microfluid handling system for further analysis.
  • This assay can also be performed by co-registering and mating a second, identical, sheet containing the fluorogenic substrate with the first sheet containing the bacteria.
  • An absorbance measurement may also be performed to normalize the signal output for the number of bacteria.
  • through-holes 12 may be disposed in an array within a circular sample wafer 320 having a central hole 322 for purposes of centering with respect to handling equipment.
  • platen 10 and through-holes 12 may assume other geometries than have heretofore been described, in accordance with alternate embodiments of the invention.
  • the volumes of the through-holes may be varied as a known function of spatial location in the array.
  • the cross section may be varied, as depicted in FIG. 5 a , as a known function of spatial location within the array by changing the hole's lateral dimensions as a function of position within the array.
  • the hole volume scales as s 2 ; thus increasing the hole dimensions by a factor of 3.3 increases the volume by a factor of 10.
  • the hole dimensions are increased as a linear function in one or both lateral directions.
  • Another method to produce different through-hole volumes as a function of array position is to change the distance between top and bottom surfaces of the plate defining the hole length 52 , as shown in FIG. 5 b .
  • One particular example is to incline the plate top 14 and bottom 16 surfaces at an angle to each other as in a wedge or prism, as shown in FIG. 5 c .
  • the volume change is linear with distance along the array in the direction of surface inclination while in the orthogonal direction along a row of holes, the hole length, and thus volume, is constant.
  • an array can be fabricated from a contiguous series of planar surfaces inclined to each other in either one- or two-dimensions such that the through-hole volume is different in both directions along the array.
  • an array is fabricated from a non-planar surface 54 such as a surface of hemispherical curvature as shown in FIG. 5 d.
  • a second array fabricated having surfaces that are the complement of the first array's geometry may be used, in accordance with alternate embodiments of the invention, to facilitates vertical stacking of the arrays, as shown in FIG. 6 .
  • Alignment of through-holes 64 of plate 60 with through-holes 66 of plate 62 is provided by inserting protruding segments 68 into corresponding indentations 70 .
  • Other array geometries with greater than four-fold rotation symmetry with respect to the array surface normal will facilitate interlocked and self-aligned stacking of arrays with matching positive and negative geometries, within the scope of the present invention.
  • This geometrical arrangement may advantageously obviate the need for alignment pins which are typically required for registration of stacked planar arrays.
  • One application of arrays having through-holes of non-uniform volume is mixing of different volumes of liquids such as in a dilution sequence, as described below.
  • An advantage of this method is that stacking of multiple arrays, as also described below, may be obviated while a wide range of dilution may be achieved.
  • the through-hole array advantageously provides the ability to collect a large number of small volume fractions, which can then be further separated within the through-hole array, as described below. This, in turn, advantageously increases the resolution of chromatographic separation over prior technology.
  • a through-hole plate 56 is employed whose thickness increases in one lateral direction (such that the plate has the shape of a prism) resulting in a linear increase in hole length with array position.
  • a second through-hole prism 57 is brought together with the first prism 56 along their common hypotenuse 58 thereby producing an array of through-holes 12 of substantially equal length.
  • each hole is filled with a porous gel 55 characteristic of liquid chromatography.
  • the sample 51 to be analyzed is applied to one end of the array, as from a sample plate 53 , for example, and a pressure is applied to drive fluid sample 51 through each hole.
  • Each component in the mixture will travel at a different velocity through the gel matrix resulting in a separation of the mixture along the column length. For example, small molecules will move rapidly through the channel to its opposite end, whereas, after the same duration of time, larger molecules may typically have traveled only a fraction of that distance.
  • the intersection of each through-hole with the hypotenuse along which the prisms are joined corresponds to different travel lengths along the chromatographic column. Separation of the array into its constituent prisms, as depicted in FIG.
  • through-holes 12 may be formed in platen 10 by any of a variety of means appropriate to the material of platen 10 .
  • Through-hole forming methods include, by way of example, laser ablation by means of an ultraviolet (UV) excimer laser which may form 100 pm through-holes in glasses and polymers.
  • Additional through-hole forming techniques include mechanical drilling, electrochemical methods such as micro-electrical spark discharge machining (EDM), employing radio-frequency pulses for ionizing removal of conductive materials, or, in accordance with other embodiments of the invention, by selective chemical or charged-particle etching techniques.
  • EDM micro-electrical spark discharge machining
  • microcapillary bundles of glass fibers of varying compositions may be drawn from preform and sliced to form platens, and then selectively etched to form through-holes.
  • through-holes 12 may be formed, in materials such as thermoplastics or polycarbide, by punching platen blank 400 using punch array 402 and die array 404 , in conjunction with a high-pressure ram.
  • Punching pins 406 may be formed in punch block 408 using microwire EDM, as described in detain in the following discussion, or by microetching techniques such as chemical or charged-particle etching.
  • punch array 402 may be formed using microsawing techniques.
  • Die array 404 is similarly formed using microfabrication techniques known in the manufacturing arts.
  • High-density arrays of through-holes may be formed in conducting materials, including conductive (>10 ⁇ ⁇ 1 -cm ⁇ 1 ) silicon wafers, using a combination of wire and die sink EDM.
  • EDM is typically used in the preparation of tooling dies for injection molding.
  • the machining process involves ionizing away the surface of a conducting material, typically a metal.
  • EDM can be performed using a tip-based electrode, or a “wire.”
  • Wire-EDM is used when fine surface finishes are required, or when subtle machining features not achievable with a tip electrode are desired.
  • Traditional wire-EDM machining utilizes wire approximately 250 ⁇ m in diameter. Due to the small dimensions and high packing densities of the invention an adapted microwire EMD process is used that employs wires with diameters down to 30 ⁇ m. This system may provide surface finishes down to 100 nm, essentially a mirror finish.
  • a typical method for preparing a through-hole array using microEDM machining follows a two-step process.
  • the first step is the creation of a positive or master die.
  • the second step uses another EDM machine called a “sink-EDM.”
  • the sink-EDM machine uses the master as an electrode and thereby creates a negative copy in the machined conducting material. This negative copy is the resulting microarray.
  • the master electrode can produce multiple negatives before needing to be replaced, thereby increasing manufacturing throughput for production of the chips.
  • the EDM technique described herein may advantageously reduce fabrication time and cost.
  • the master die After the master die is fabricated with wire EDM, it may typically be used to sink an array of microchannels through a 0.5-mm thick silicon wafer in less than two minutes. Additional machining time, during which the master die moves largely in the plane of the array, is typically needed to enlarge the holes to the desired dimension and to improve the surface finish.
  • the fabrication process described has been used to sink microchannels of sizes up to 10 000 elements with cross-sections ⁇ 300 ⁇ m ⁇ 300 ⁇ m and element spacings ⁇ 500 ⁇ m. Another advantage of this technique is that it may also be used to a manufacture stainless steel precision alignment jig that is used to align the chips for mixing and optical readout, as described herein.
  • this EDM process results in ⁇ 10 ⁇ m center-to-center total error in channel spacing across the entire array. This guarantees accurate hole alignment across arrays, and when combined with the hydrophobic exterior coating, minimizes cross talk between microchannels.
  • the master die 408 produced from the process outlined above can be used to create the through-hole arrays from a plastic blank 400 .
  • the die is used as a type of punch to create the through-hole array from a solid piece of plastic.
  • the master does not force its way through the plastic. Instead, by having the appropriate kinetic energy as it impacts the plastic it essentially vaporizes the solid plastic into low molecular weight gases (with some residual energy dissipated as heat). In this manner the master can be removed from the resulting through-holes without having melted into it. A slight taper on the protrusions of the master that forms the channels facilitates removal of the master from the chip.
  • This process is similar to that which is used in the manufacturing of DVD's, except the manufacture of microwell arrays typically requires significantly greater penetration depths (up to 1 mm deep).
  • plastic microwell plates may be manufactured by injection molding of metal masters formed by EDM.
  • the inner coating 24 may be chemically blocked to prevent non-specific binding or derivatized with affinity ligands.
  • a dense array 10 of through-holes 12 is produced in silicon and coated in silicon oxide by oxidation.
  • the surfaces of the conductive silicon are covered in a thin oxide layer.
  • the wafer is then cleaned by soaking in a mixture of hydrogen peroxide and sulfuric acid, or other caustic solvent cleaning solution, to remove organic materials. Clean silicon oxide thus produced has a high surface energy.
  • the top and bottom faces of the arrays are made hydrophobic by exposing them to vapor from a solution containing an appropriate silanizing agent (such as polydimethylsiloxane, sold as Glassclad 216TM by United Chemical Technologies, Inc.) in a volatile solvent.
  • the silanizing agent reacts with the hydroxyl and silanol groups on the array surface and creates covalent bonds to hydrophobic alkyl groups.
  • the resulting coated arrays can be uniformly loaded with aqueous solutions by simply dipping the array into it.
  • the liquid instantaneously fills the channels through capillary pressure, but does not wet the other surfaces.
  • Hydrophobic coatings produced in this way are stable under high humidity and they can be used repeatedly over several days.
  • Other surface chemistries may be exploited to attach hydrophobic chemical groups to the faces of arrays made from other materials. For example a gold-coated surface reacts with alkane thiols to attach a hydrophobic alkyl groups, as discussed by C. D. Bain et al., J. Am. Chem. Soc ., vol. 111, pp. 321-325 (1989), which reference is incorporated herein by reference.
  • the surface chemistry of the array faces may also be selectively modified by substituting other silanizing agents for the polydimethylsiloxane telomer.
  • This method may advantageously prevent aqueous solutions from adhering to array faces during array loading and also act as physical barriers between the aqueous solutions in adjacent through-holes. During this process a positive pressure of inert gas is applied to the opposite side of the array. The positive pressure within the through-holes prevents the silanizing vapor from reaching the interior surfaces. This method advantageously allows removal and reaplication of the hydrophobic coating.
  • array 10 of through-holes 12 may be filled using techniques either that directly address particular through-holes, or techniques that fill the entirety of the array according to a specified pattern based on composition, concentration of a substance, etc.
  • Dip loading may be employed, for example, in order to fill an entire array with the same solution.
  • a through-hole array 10 is produced and chemically treated to make the array faces hydrophobic and the through-hole surfaces hydrophilic.
  • Plate 10 with through-holes 12 is first lowered into a container 90 containing a first liquid 82 , as shown in FIG. 9 a . Once plate 10 is fully immersed in the first liquid, it is shaken so that first liquid 82 displaces the air in each of the through-holes, and all the through-holes 12 are filled with first liquid 82 .
  • plate 10 is withdrawn from first liquid 82 , as shown in FIG. 9 c .
  • the array is typically withdrawn slowly such that the surface of the fluid in the reservoir pulls excess liquid off of the non-wetting array surface.
  • the array may be filled with water by spraying the array with water or by sonicating the array in a reservoir of water in order to remove trapped air bubble prior to dip loading. In this case, less agitation is required to uniformly fill the array with solution.
  • Both convection and diffusion can be used to induce mixing between fluids in microchannel arrays. This can be demonstrated by filling a microarray with aqueous solution of blue dye and by submerging the microarray into a beaker of still water to induce dilution via diffusion. A small mechanical disturbance (such as a tap to the side of the beaker) causes rapid replacement of the blue dye by water.
  • Alternative methods for filling the array with water include spraying the array with water or sonicating the array in a reservoir of water in order to remove trapped air bubble prior to dip loading are also within the scope of the present invention.
  • an array having hydrophilic faces and hydrophobic through-holes may be uniformly filled with a low surface tension liquid such as an alkane.
  • Droplet dragging is performed to load an entire array or a group of channels within a single array with the same fluid. It is appropriate for loading samples which must be conserved because of cost or availability.
  • a droplet of the loading solution is formed on the end of a syringe needle, micropipette or other fluid dispensing capillary.
  • the drop is placed on the array face over the through-holes to be filled.
  • the capillary is then moved to drag the drop across the surface of the array and over the channels to be filled. Surface tension maintains contact between the capillary tip and the fluid drop.
  • additional fluid is dispensed until all desired through-holes have been filled.
  • the capillary and any remaining fluid in the drop are then withdrawn from the array surface.
  • a method is provided to transfer liquids from standard microtiter plate formats (96-, 384-, or 1536-wells, for example) to a single array of through-holes or multiple, vertically stacked arrays in which holes in the same spatial location from one array to the next are co-registered.
  • registration refers to aligning a through-hole plate with at least one other through-hole plate, such that the tops of a plurality of through-holes of one of the plates coincide with the bottoms of a plurality of corresponding through-holes of the other plate or plates, thereby creating a plurality of contiguous channels.
  • All of the filling examples given can be performed in parallel on a single array stack with multiple capillary tubing arrays filled with liquids from different microtiter plates or the same microtiter plate. Fluid may be transferred from chemical libraries stored in 96- and 384-well microtiter plates quite rapidly with these methods. For example, if 100 arrays with 10 5 through-holes per array are to be filled with liquid stored in 96 well plates, and if each transfer operation (plate exchange, fluid loading and transfer) takes approximately 20 s, then the requisite arrays may be filled within approximately six hours. The filling operations take place preferably in an environmentally-controlled chamber (temperature, humidity, atmosphere, etc.), as described below.
  • FIGS. 10 and 11 further embodiments of the invention provide an apparatus and methods that are particularly suited to the transfer of liquids from standard microtiter plate formats (such as 96-, 384-, or 1,536-well formats) to a single array of through-holes or multiple, vertically stacked arrays in which holes in the same spatial location from one array to the next are co-registered. Producing such stacks of arrays may be employed advantageously for producing replicates of molecular or cellular libraries.
  • standard microtiter plate formats such as 96-, 384-, or 1,536-well formats
  • a capillary tube array 100 is constructed from capillary tubing 102 with an external diameter that fits precisely into the through-holes of a through-hole microwell array 10 .
  • Tubing array 100 is designed such that tubing 102 at one end 104 has a center-to-center spacing 106 equal to the spacing between holes in through-hole array 10 (or, alternatively, to an integral multiple of the inter-hole spacing) and tubing at the opposite end 108 has a center-to-center spacing equal to the center-to-center spacing 110 of wells 112 in a microtiter plate 114 .
  • Plates 116 , 118 , and 120 with through-holes having these and intermediate separations serve as jigs to hold the tubing in a regular array. Additional through-hole plates 116 , 118 , and 120 placed between the two ends may advantageously serve as spacer jigs providing additional support for the tubing array as the center-to-center spacing varies over the tubing length.
  • each tube 102 in the capillary array 100 is slightly greater than the total volume of a column of aligned holes in the array stack 122 .
  • the volume of one set of holes in a stack of 100 arrays is 6.25 ⁇ l (100 ⁇ 62.5 nl).
  • Capillary tubing with an internal diameter of 200 ⁇ m and an external diameter of 245 ⁇ m is readily available; thus a minimum tube length of 200 mm stores the volume of fluid needed to fill this set of through-holes.
  • tubing array 100 is inserted into the wells of a microliter plate 114 , each tube being inserted into a corresponding well 112 .
  • a negative pressure difference is applied across the length of tubing 102 to draw liquid, in direction 126 , from each well into its corresponding tube. Negative pressure could be applied to each tube individually, or as shown in FIG. 10 , the ends of the tube array can terminate in a chamber 124 that can be partially evacuated.
  • the microtiter plate 114 is removed.
  • the liquid can be stored in the tubing array for an indefinite period of time, either frozen or in a humidified environment. It can also be readily transported to another location in this format. Multiple tubing arrays can be filled from the same microtiter plate (assuming there is sufficient volume of liquid per well) or different tubing arrays can be filled from different microtiter plates.
  • Embodiments of the invention also provide for methods for filling a stack of arrays, as shown in cross-section in FIG. 11 , by bringing the end of a tubing array 130 into close proximity with a matching set of through-holes in the array stack 122 .
  • the tubing array can be aligned relative to the array stack by an alignment plate 128 with through-holes having the same center-to-center spacing as the through-holes into which fluid is placed.
  • Each array 10 in the stack 122 is spaced a small distance s that may be, but is not limited to, an equal distance to the through-hole spacing.
  • Application of pressure to the end of the tubing array, placed inside a pressurized container 132 forces fluid from each capillary tube 102 into the opposing through-hole.
  • a liquid drop can begin to grow in the space between the two plates.
  • the drop reaches a size that it contacts the through-hole in the plate above it, surface tension draws some fluid into the through-hole.
  • liquid can flow into the through-hole, driven by the constant pressure applied to the opposite end of the tubing array. With no applied pressure, the drop retreats into the through-hole, the fluidic bridge between each plate is broken, and the separation of array plates after filling can be facilitated (i.e., because there is generally no surface tension that needs to be overcome).
  • Successive filled plates 10 are then withdrawn, and the tube array may be retracted in direction 128 .
  • Each vertically registered set of through-holes may thus act as a channel for fluid flow.
  • FIG. 12 A method for filling a through-hole array stack, in accordance with alternate embodiments of the invention, is shown in FIG. 12 .
  • This method uses a through-hole array 10 having the same lateral dimensions as a microtiter plate 114 and having a hole spacing that is an integral fraction of the well spacing in the microtiter plate 114 .
  • the array is placed on top of the plate, one or more through-holes 12 align with respect to each well 112 in microtiter plate 114 .
  • An array 130 of syringes 132 with a center-to-center spacing equal to the well spacing can thus be positioned over a stack of through-holes registered with respect to each other and the microtiter plate.
  • the syringe array is inserted through a through-hole plate 134 , such that the plate is a mechanical guide for the syringe tubing as it is moved relative to the array stack.
  • the syringe plungers are mechanically coupled and actuated by a mechanical or electromechanical driver module 136 , such that liquid is drawn into or expelled from each syringe in parallel.
  • the syringe tubing outside diameter is preferably sized relative to the through-hole lateral dimensions to give a sliding fit, and the tubing can have a length suitable to allow insertion through the array stack 122 and into the liquid contained in the microtiter plate wells 112 .
  • the volume of liquid withdrawn into each syringe preferably equals the volume of liquid in a column of aligned through-holes in the array stack.
  • the liquid can then be dispensed as the syringe array is retracted from the array stack, and the rate of dispensing can be synchronized with the rate of withdrawal, such that each through-hole addressed by the syringe array is filled.
  • a new set of holes in the array stack can be filled from the same microtiter plate or the syringe array can be washed and different set of holes filled from a different microtiter plate.
  • All of the filling examples given above may be performed in parallel on a single array stack with multiple capillary tubing arrays filled with liquids from different microtiter plates or the same microtiter plate.
  • the time to transfer fluid from chemical libraries stored in 96 and 384 well microtiter plates with these methods can be quite rapid. Assuming, for example, that 100 arrays with 10,000 through-holes per array are to be filled with liquid stored in 96 well plates, and that each transfer operation (plate exchange, fluid loading and transfer) takes approximately 20 seconds, then it would take approximately six hours to fill all of the arrays.
  • the filling operations can take place in an environmental controlled chamber (temperature, humidity, atmosphere, etc.).
  • the invention also provides a method for screening compound libraries to predict the ability of each compound to be absorbed by a patient.
  • fluid can be transferred from individual wells 112 of a microtiter plate 114 with an array 140 of flexible members 142 , e.g., shape memory alloy fibers.
  • the fiber diameter is equal to or less than the inside dimension of the through-holes 12 in the array 10 into which fluid will be transferred.
  • the number of fibers in the bundle may, for example, be equal to the number of wells in the microtiter plate 114 .
  • the ends of the fibers at one end of the bundle can have a center-to-center spacing equal to the spacing of the holes in the through-hole array, while the ends of the fibers at the opposite end can have a center-to-center spacing equal to the spacing of wells in the microtiter plate.
  • the fibers can be held in place with a series of through-hole jigs designed to increase the spacing between fibers from one end of the bundle to another. Once fixed in place, shape memory alloy fibers can be heated above their critical transition temperature to make the imposed fiber curvature permanent. After they are cooled to room temperature, the fibers can be removed from the holding jig, with the change in fiber center-to-center spacing intact.
  • the close packed end of the fiber bundle can then be inserted into the through-hole array into which fluid from each well in the plate is to be placed.
  • the opposite end can be arranged such that each fiber is positioned above a well in the microtiter plate, and the ends of the fibers can be immersed in the fluid contained in each well.
  • a small volume drop 144 remains attached (e.g., by surface tension) to the end of each fiber 142 .
  • a force may be applied to the opposite end of the fiber bundle to pull the bundle through the holes 12 of the through-hole array 10 , such that the fluid is brought into contact with the corresponding through-holes.
  • surface tension acts to hold the liquid in the through-hole as the fiber is removed.
  • Addressable loading may also be employed to fill a series of through-holes with different and specified concentrations of the same solute.
  • a chosen series of through-holes is filled with a quantity of solvent, denoted Z nanoliters (nL), either by dipping the holes into the solvent or by dispensing the solvent from a microsyringe 232 (shown in FIG. 21 ), or other fluid transfer device. This is shown as the first step 120 in the flow chart of FIG. 13 .
  • microsyringe 232 or another fluid transfer device, is filled with an X molar solution of the solute, and is then positioned over the first through-hole.
  • Y nL of this solution where (Y+Z) nL is sufficient volume to overfill the hole and to create positive menisci, is expelled such that it forms a droplet at the end of the syringe tip.
  • the syringe tip is lowered until the solvent droplet contacts the surface of the solution, causing the two liquids to mix 122 and produce a solution of concentration YX/(Y+Z) molar.
  • the outer surface of the syringe tip and the faces of the array must be nonwetting toward the solution being dispensed.
  • the syringe plunger is then withdrawn in step 123 to suck up Y nL of the diluted solution.
  • the syringe tip is positioned above the next (N+1) through-hole 124 and Y nL of the diluted solution is dispensed into the solvent to dilute 125 by another factor of Y/(Y+Z).
  • the process is repeated so as to dispense solution into a series of individual through-holes, each time diluting by Y/(Y+Z).
  • a particular chemical species is not loaded uniformly into all the holes of an array, but, rather, a gradient of chemical species is created in at least one dimension in a two-dimensional through-hole array.
  • the term “gradient” refers to its ordinary mathematical meaning, i.e., a variation, along one or more directions, of a specified quantity. The directions, here, are taken along the surface of an array of through-holes.
  • a gradient in the concentration of a specified chemical species in the through-holes may be said to exist with respect to a particular direction, or, for that matter, with respect to various directions.
  • a particular gradient may be, but need not be, monotonic, and the concentration of a species, in a specified direction, may rise and fall.
  • dip loading methods described above may be used for creating a concentration gradient in an array of through-holes.
  • the array is dipped into a chemical solution at a controlled rate such that the chemicals in the solution have different amounts of time to mix with, or react with, substances by diffusion into various through-holes.
  • plate 10 is subsequently lowered into a second container 84 containing a second liquid 86 , as shown in FIG. 14 a . Since the through-holes of plate 10 are now filled with the first liquid 84 , the second liquid 86 may diffuse into, mix with, displace, or otherwise react with, the contents of through-holes 12 . Each of these processes is characterized by a rate. Instead of fully immersing the plate until the particular reaction, physical or chemical, runs to completion, plate 10 may be partially immersed into second liquid 86 , as shown in FIG.
  • FIG. 14 d a side view is shown of through-hole array plate 10 immersed in liquid 86 . Diffusion of liquid 86 into through-holes 12 is modulated by membrane or filter 87 , for example, that, by virtue of its tapered shape or otherwise, gives rise to a gradient of a specified species with respect to the position of a through-hole within the array.
  • concentration gradients within through-hole arrays are use to optimize the concentrations of A and B in a two-component reaction between Reagent A and Reagent B.
  • the mixing process is described in greater detail below.
  • a first through-hole array shown in . cross section and designated generally by numeral 90 , is loaded with Reagent A using a chemical gradient method that results in the through-holes along one row, in the x direction, of the array will have the same concentration of A and the concentration gradient is along the array columns, in the z direction.
  • a second array designated generally by numeral 92 , is loaded with Reagent B such that the concentration gradient is along the rows (x direction) of the array and through-holes along a column (z direction) of the array have all the same concentration. Bringing the two arrays in contact causes mixing between aligned through-holes 12 . Along each row or column of the combined arrays the concentration of A relative to B in the mixture changes in a regular and known manner. Along the array diagonal, the concentrations of A and B in the mixed liquids are equal and but changing in overall concentration.
  • Variants from the basic scheme for creating a known two-dimensional gradient in the relative proportions of A and B include loading the A and B arrays by dipping along a diagonal, by the orthogonal combination of two arrays each with a 2-D gradient of A relative to B or by dilution with arrays loaded with Reagent A or B where some of the through-holes are empty. Analysis of the through-hole contents readily determines optimal reaction conditions since the reaction conditions in each through-hole are known. Gradients created in the solution with respect to quantities other than solute concentration are also to be understood to be within the scope of the present invention, and may include, without limitation, such characteristics as temperature, electric field, magnetic field, etc.
  • microwell plates may advantageously be stacked for such purposes as mixing or dilution.
  • One such application is the apportionment among through-holes of a sample of cells. Considering, for purposes of example only, through-holes on a single plate that are 250 ⁇ m square and 500 ⁇ m deep.
  • the total volume of a single channel i.e., the combined volume of three through-holes
  • the entire channel is filled from a dense yeast cell culture ( ⁇ 10 7 /mL) each channel then contains approximately 10 3 yeast cells.
  • the maximum number of cells per 100 nL channel is on the order of 10 6 , consonant with a typical minimum of 100 cells per microchannel is required to compensate for variable yeast cell response to the bioassays.
  • FIGS. 15 -19 mixing and dilution are shown between the contents of two substantially planar through-holes array plates 90 , 92 (i.e., array plates having neither flanges nor indentations).
  • FIG. 15 a cross-sectional view is shown of portions of two through-hole arrays 90 , 92 .
  • Both the top and bottom surfaces of each platen are hydrophobic, as discussed in detail above.
  • the through-hole walls 24 are preferably hydrophilic.
  • Through-holes 12 of both platens are overfilled with high surface energy fluids 150 , such as aqueous solutions, for example, such that each through-hole 12 has a positively-curved meniscus 152 protruding above the platen surface 154 .
  • the platen surfaces 154 may be hydrophilic and a sufficient amount of a low surface energy fluid, such as an alkane, for example, is loaded into the through-holes 12 to form positive menisci 152 .
  • a low surface energy fluid such as an alkane
  • FIG. 16 shows a cross-sectional view of the two platens 90 , 92 after the surfaces of the liquids 150 in co-registered through-holes 12 are brought into contact.
  • the release of the surface tension drives convective mixing between fluids contained in the opposite platens 90 , 92 .
  • each set of two-coregistered through-holes forms a longer channel 170 , as shown in FIG. 17 .
  • Mixing proceeds within the longer channels by a combination of convection and statistical diffusion.
  • first platen 90 is filled with sufficient fluid 150 to form positive menisci 190 , 192
  • a second platen 92 is filled with an amount of fluid 194 that is insufficient to form positive menisci, but sufficient to make contact with the surface 190 of the menisci of the first platen when the two platens are stacked.
  • Mixing proceeds by bringing the liquid surfaces into contact as previously described.
  • a level of humidity required to keep the volume loss at an acceptable level depends upon the length of time needed to perform the various desired operations, as well as on such factors as the ambient temperature, and the volume of fluid contained in each through-hole.
  • the evaporative loss expected from a droplet of water may be predicted in accordance with Fick's law, as depicted in FIG. 20 .
  • a through-hole array 10 is mounted above a reservoir 210 of water.
  • a 96-well, 384-well or higher density microtiter plate 212 is mounted above the same reservoir proximal to the through-hole array.
  • Reservoir 210 may contain a water absorbent material such as a sponge 214 to keep the water from sloshing when the reservoir is moved.
  • An optically transparent plate 216 is placed on top of the reservoir separated from the walls of the reservoir by a thin layer 218 of a viscous non-hydroscopic fluid such as silicone grease, for example.
  • One or more small holes 220 are drilled through transparent plate 216 . Through each hole a microsyringe needle 222 , microcapillary, pin, cannula, or other fluid transfer element is inserted.
  • Fluid transfer element 224 may include syringe 232 with plunger 234 fluid transfer element 224 , for example. Additionally, the arrays and reservoir may be moved in the plane of the x and y axes, preferably by motorized stage 228 under control of controller 230 , with respect to the fluid transfer element. If indicated, further computer-controlled stages may be provided, for example, to actuate the fluid transfer device. Transfer of materials between the microtiter plate and through-hole array proceed by means described above in reference to FIGS. 10-12 .
  • Additional reservoir(s) containing solution(s) for cleaning/sterilizing the fluid transfer element may also be provided within the confines of chamber 236 which encloses the apparatus heretofore described. Alternatively the water in the large reservoir 210 may be used for this purpose.
  • An illumination source 238 may also be provided for to illuminate the array for optical inspection during loading/unloading, either visually or by means of a video camera 240 .
  • An environment enclosure 236 may also be utilized in order to preventing evaporation from the through-hole arrays during dip loading and mixing.
  • Sealable chamber 236 is large enough to contain the array and necessary apparatus for performing such desired operations as are required, including, for example, motors and translation stages 228 , as well as an alignment jig 156 (shown in FIG. 15 ) for mixing operations.
  • Cool water vapor is generated by an external humidifier and injected into the chamber through a port.
  • An ultrasonic humidifier is able to generate enough vapor to maintain humidity levels above 95% in a 0.13 cubic meter chamber.
  • a circulation system consisting of a fan and baffles is provided to distribute the water vapor uniformly throughout the chamber.
  • Various doors, hatches and iris ports may be provided for accessing the interior of the chamber.
  • the temperature within the box may be controlled by a resistive heater and an electronic temperature controller.
  • Such a chamber may be assembled by attaching a second humidifier to a commercial infant incubator.
  • a humidity sensor is also provided to monitor humidity levels inside the chamber. All computer and electronic hardware are placed outside the chamber and are connected to components inside the chamber via a wire feedthrough.
  • the through-hole array may be filled within the chamber, or loaded externally and transferred into to the chamber in a humidified sealed cassette.
  • FIG. 22 a an exploded perspective view is provided of a compact portable cassette, designated generally by numeral 250 , for maintaining relative humidity levels above 95% when the array is removed, for optical analysis, for example, from the humidified loading chamber 236 (shown in FIG. 21 ).
  • FIG. 22 b provides a side view, in cross section, of the humidified cassette 250 of FIG. 22 a .
  • Through-hole array 10 and an aqueous microlens array 252 are mounted in a compact, sealable enclosure.
  • the though-hole array is positioned and held in place by means of alignment pins 256 or some other means.
  • Both the array mount 258 and cover holder 260 are covered by an optically transparent plate 262 secured to the mount and cover by adhesive or other mechanical means.
  • the mount may also include magnets, pins, grooves or other physical features to facilitate position the array inside optical analysis equipment.
  • the humidity is raised by passive evaporation from water contained in a transparent glass microcapillary bundle 252 (or, ‘microchannel array’) mounted beneath the through-hole array and secured by set screws or some other mechanical means.
  • Glass microcapillary bundles are manufactured by Schott Fiber Optics, Southbridge, Mass.).
  • Typical dimensions for each capillary 264 are diameter of 200 microns and depth of 1 mm.
  • the top 266 and bottom 268 of the microchannel array 252 are made hydrophobic according to the procedures described above.
  • the arrangement of the capillaries 264 in the array need not be regular.
  • Water 270 in the capillaries forms a set of microlenses which serve to diffuse light from light source 272 across the array and thus provide a uniform illumination field for optical analysis.
  • the use of liquid microlens arrays generally, is within the scope of the present invention.
  • aqueous lens array Another advantage of the aqueous lens array is that the water is held in place by surface tension. Thus the operator need not keep the humidity cassette level, or avoid applying the typical accelerations that occur when the plate is carried around a room and placed on horizontal surfaces. Also the cassette may be agitated during incubation to promote cell growth inside the arrays.
  • Pressure, light and temperature, are controlled by enclosing the array in an appropriately constructed chamber and then controlling the environment inside the chamber by conventional means. To prevent evaporation such chamber must also be equipped to maintain high relative humidity.

Abstract

A method and an array filling system for loading a plurality of disparate sample containers, the sample containers comprising an integral structure. Each receptacle is characterized by a hydrophilic surface,, and the receptacles are separated by a hydrophobic surface. The system has a liquid transfer device capable of holding liquid and adapted for motion to cause sequential communication of liquid held in the liquid transfer device with successive receptacles of the array by dragging the liquid across the hydrophobic surface.

Description

  • The present application is a divisional of copending U.S. patent application Ser. No. 10/820,679, filed Apr. 8, 2004, itself a divisional of U.S. Ser. No. 09/850,123, filed May 7, 2001, claiming priority from U.S. Provisional Application No. 60/239,538, filed Oct. 10, 2000, and also a continuation-in-part of U.S. Ser. No. 09/225,583, filed Jan. 5, 1999, now issued which in turn claims priority from U.S. Provisional Application No. 60/071,179, filed Jan. 12, 1998, from all of which applications the present application claims priority, and all of which applications are herein incorporated by reference.
  • FIELD OF THE INVENTION
  • The present invention pertains to methods for manufacturing and using apparatus for manipulating, transporting, and analyzing a large number of microscopic samples of a liquid or of materials including cells currently or formerly in liquid suspension.
  • BACKGROUND OF THE INVENTION
  • Chemistry on the micro-scale, involving the reaction and subsequent analysis of quantities of reagents or analytes of order microliters or smaller, is an increasingly important aspect of the development of new substances in the pharmaceutical and other industries. Such reaction and analysis may accommodate vast libraries containing as many as a million compounds to be reacted and analyzed under various conditions. Significant problems associated with current technologies as applied to chemical analysis of vast numbers (potentially on the order of hundreds of thousands or millions per day) of compounds include the problem of handling vast numbers of compounds and reactions in parallel.
  • Existing technology relies on 96-, 384-, or 1536-well plates containing quantities between approximately 1 microliter and 1 milliliter of liquid compound per well, and, generally, involves chemical reactions and analysis in wells disposed with single openings on flat surfaces such as polystyrene. It is not practical to apply existing technology in the art to form million-well microtiter plates. There is a need, therefore, for new approaches that permit the analysis of a million samples in a laboratory format.
  • SUMMARY OF THE INVENTION
  • In accordance with preferred embodiments of the present invention, a method and an array filling system are provided for loading a plurality of disparate sample containers, the sample containers comprising an integral structure. In the array filling system for transferring liquid to an array of receptacles, each receptacle is characterized by a hydrophilic surface, and the receptacles are separated by a hydrophobic surface. The system has a liquid transfer device capable of holding liquid and adapted for motion to cause sequential communication of liquid held in the liquid transfer device with successive receptacles of the array by dragging the liquid across the hydrophobic surface.
  • In accordance with other embodiments of the invention, the array filling system has an array of liquid transfer devices capable of holding liquid, the plurality of liquid transfer devices adapted for motion to cause sequential communication of liquid held in the liquid transfer devices with successive receptacles of the array by dragging the liquid across the hydrophobic surface.
  • In accordance with yet other embodiments of the invention, a method is provided for transferring a liquid into an array of receptacles having hydrophilic surfaces and separated by a hydrophobic surface. The method has steps of:
      • a. positioning a liquid transfer device in proximity to the surface; and
      • b. moving the fluid transfer device across the hydrophobic surface and over the receptacles to be filled so as to cause sequential communication of liquid in the liquid transfer device with the hydrophilic surface of each receptacle.
  • In further embodiments of the invention, the method may have a further step of withdrawing residual fluid from the surface of the platen. The step of causing communication of liquid in the liquid transfer device with the hydrophilic surfaces of the plurality of hydrophilic receptacles may include forming a droplet of the liquid on the end of a capillary, or on the end of a pipette or needle, and may, further, include transferring liquid into through-holes of an array of through-holes.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • The invention will be more readily understood by reference to the following description, taken with the accompanying drawings, in which:
  • FIGS. 1 a and 1 b show a top and exploded cross-sectional views, respectively, of a high-density array of through-holes in accordance with one embodiment of the present invention;
  • FIG. 1 c shows a schematic side view of a continuous sheet array of through-holes in accordance with another embodiment of the present invention;
  • FIG. 2 a is top view of a portion of the platen of FIG. 1 a in which the through-holes are configured on rectangular centers;
  • FIG. 2 b is top view of a portion of the platen of FIG. 1 a in which the through-holes are configured in a hexagonal close-packed array;
  • FIG. 3 is a further side view in cross-section of a portion of a laminated platen containing multiple through-holes for analysis of liquid samples in accordance with a preferred embodiment of the present invention;
  • FIG. 4 is a top view of round sample wafer populated with through-holes in accordance with an embodiment of the present invention;
  • FIGS. 5 a-5 d show examples of arrays with through-hole volumes that are a function of array position;
  • FIGS. 5 e and 5 f show cross sections of through-hole prisms as employed for massively parallel liquid chromatography or electrophoresis, in accordance with embodiments of the present invention;
  • FIG. 6 shows an example of an interlocking array;
  • FIG. 7 depicts the configuration of a compression mold for compression molding on an array of through-holes in accordance with an embodiment of the present invention;
  • FIG. 8 is a flow chart depicting steps in the fabrication of a through-hole array by EDM, in accordance with an embodiment of the invention;
  • FIGS. 9 a-9 c depict a sequence of operations for filling a through-hole array with a first liquid;
  • FIG. 10 is a schematic depiction of a method for sampling the contents of a large-format microtiter plate and loading a subarray of the through-hole array in accordance with an embodiment of the present invention;
  • FIG. 11 is a schematic depiction of a method for sampling the contents of a large-format microtiter plate and loading subarrays of multiple through-hole arrays in accordance with an embodiment of the present invention;
  • FIG. 12 a depicts parallel loading of subarrays of multiple stacked microwell arrays in accordance with embodiments of the present invention;
  • FIGS. 12 b and 12 c depict parallel loading of subarrays of a microwell array by means of flexible members, in accordance with embodiments of the present invention;
  • FIG. 13 is a flow chart depicting a method for successive dilution of the contents of a microwell array in accordance with an embodiment of the present invention;
  • FIGS. 14 a-14 c depict a sequence of operations for exposing respective through-holes of a through-hole array to a second liquid and for creating a specified gradient of a specified characteristic with respect to placement of the through-holes in the array in accordance with an embodiment of the present invention;
  • FIG. 14 d shows a side view is shown of a through-hole array plate with a graduated filter for providing a concentration gradient in accordance with an embodiment of the invention;
  • FIG. 15 depicts mixing of the contents of a through-hole array with the contents of another through-hole array brought into registration of the through-holes in accordance with embodiments of the present invention;
  • FIGS. 16 and 17 depict further stages in the mixing of the contents of a through-hole array with the contents of another through-hole array brought into registration of the through-holes in accordance with embodiments of the present invention;
  • FIG. 18 depicts mixing of the contents of a through-hole array with the contents of another through-hole array by application of external pressure, in accordance with embodiments of the present invention;
  • FIG. 19 depicts mixing of the contents of a fully filled through-hole array with the contents of another incompletely filled through-hole array brought into registration of the through-holes in accordance with embodiments of the present invention;
  • FIG. 20 shows plots of calculated times for evaporation of a water droplet as a function of the ambient relative humidity;
  • FIG. 21 depicts a humidity chamber for loading and unloading through-holes while maintaining a high relative humidity in the environment surrounding the array, in accordance with an embodiment of the present invention;
  • FIG. 22 a depicts an exploded perspective view of a portable humidity chamber for preventing evaporation of fluid from the arrays during fluorescent imaging analysis, incubation and transferring the array between other humidified environments; and
  • FIG. 22 b is a cross-sectional side view of the a portable humidity chamber of FIG. 22 a.
  • DETAILED DESCRIPTION OF SPECIFIC EMBODIMENTS The Through-Hole Array
  • In accordance with the present invention, methods are provided for producing different chemical reactions within an array of through-holes. The invention is advantageously employed, for example, in screening operations, where different reaction conditions are advantageously provided among the various through-holes of the array. As an example of the many modalities of use of the invention, different chemical species may be loaded into different through-holes of the array, and concentrations of the various species might also be differentiated among the various through-holes. The invention may thus provide a method for screening compound libraries, for example, to predict the ability of each compound to be absorbed by a patient.
  • In accordance with preferred embodiments of the invention, a high-density array of through-holes is provided, as now discussed with reference to FIGS. 1 a and 1 b. FIG. 1 a shows a top view of a platen 10, otherwise referred to herein as a “substrate,” “sample wafer,” or “through-hole plate.” Platen 10 is the carrier of a large number of through-holes 12 which traverse platen 10 from a top surface 14 to an opposing surface 16 of the platen, as shown in the cross-sectional side view of FIG. 1 b. While the term “platen” may refer to a structure having substantially parallel plane surfaces and transverse dimensions substantially exceeding the thickness of the structure, alternative geometries are within the scope of the present invention, and use of the term “platen” is not restrictive. A prism-shaped geometry is described below, for example.
  • Through-holes 12 constitute assay wells (or “microwells”) in accordance with an embodiment of the invention. Through-holes 12 may be shaped as circular right cylinders, or, alternatively, may have rectangular cross-sections. Otherwise-shaped through-holes are also within the scope of the present invention.
  • While through-hole plate 10 is preferably made of conductive silicon, other types of rigid materials, such as metal, glass, or plastic may be used provided that the material is chemically inert with respect to the sample substances, or can be rendered so by appropriate surface treatments.
  • Each through-hole 12 is typically of a substantially square cross-section, although geometries, such as circular or rectangular cross-sections may be used. Through-holes 12 are also referred to herein as “channels.”
  • Through-holes 12 may be centered on a rectangular grid, as shown in FIG. 2 a, or in a close-packed hexagonal lattice, as shown in FIG. 2 b. Referring to FIG. 3, a typical thickness 20 of platen 10 is on the order of 0.5-2 mm, while through-holes 12 have typical characteristic dimensions (such as diameters) 36 of on the order of 100-400 μm. Thus the volume of each through-hole 12 between surface 14 and surface 16 is on the order of ˜10−7 cm3 or greater. Through-holes 12 are spaced on centers typically on the order of twice the diameter of the holes, although all spacing configurations are within the scope of the invention and of the appended claims. In particular, a hole-to-hole spacing 38 of 500 μm is typically employed, which corresponds to an array density of 400 holes per square centimeter of plate. In accordance with manufacturing methods described below, microwells are produced for the assay of a chemical or biochemical reaction where the volume of each microwell may be less than 100 nanoliters (10−7 cm3). The packing density of wells may thereby be increased by several orders of magnitude over prior art technology.
  • Grouping of through-holes into smaller sub-arrays may also be used, and, more particularly, a reproducible pattern may be applied to a plurality of the sub-arrays. Each through-hole 12 may be identified by its own address within the array.
  • Referring, again, to FIG. 1 b, platen 10 may also advantageously be formed of a laminate of materials, with a central layer 24 and outer “sandwiching” layers 22. In order to enhance capillary loading of sample 30 into the microwell and to prevent capillary outmigration of the sample liquid, exterior sections 22 of the microwell, adjacent to surfaces 14 and 16 of platen 10, have a hydrophobic wall surface in accordance with a preferred embodiment of the invention, while the interior section 24 of the through-hole wall has a hydrophilic surface thereby preferentially attracting an aqueous liquid sample. In a similar manner, an array having hydrophilic faces and hydrophobic through-holes may be uniformly filled with a low surface tension liquid such as an alkane. The hydrophobic layers on either end of the well are on the order of 1 μm thick or less. On loading the sample liquid into the microwells, each well is typically overfilled by about 10% above the volume surrounded by the four walls of the microwell. Under these circumstances, liquid sample 30 may form convex meniscus surfaces 34 on both the upper and lower surfaces of the sample.
  • An underfilled microwell 26 will typically be characterized by a liquid sample exhibiting a concave meniscus 28 on both the upper and lower surfaces of liquid sample 30.
  • The apertures of through-holes 12 need not be square, and, in accordance with an alternate embodiment of the present invention, flanges 8 may extend above planar surface 14 surrounding some or all of through-holes 12 while indentations 6 may be fabricated rounding the edges of through-holes 12 at opposing surface 16. Flanges 8 and indentations 6 may advantageously provide for registration of successive platens 10, in the case where platens are stacked, and in processes of mixing or dilution, as discussed in detail below.
  • Through-holes 12 may be loaded with a first sample 18 in liquid form. Sample 18 is allowed to react with a second sample where the second sample may include a variety of test samples and by subsequent or concurrent analysis of the reaction products, using, for example, optical markers, a large number of reactions may be processed and analyzed in parallel.
  • As applied to biological assays, by way of example, first sample 18 may be a solution containing pharmacologically relevant proteins or other molecules. Such a solution may include, for example, cells in aqueous suspension, eukaryotic (animal, yeast) or prokaryotic (bacteria) cells, hybrid cells, and biological molecules including, for example, antibodies and enzymes, although application to other biological or non-biological assays is within the scope of the invention as claimed herein. All such reagents may also be referred to herein and in the appended claims as “targets.” Typical yeast cell concentrations of 107 cells per milliliter of solution yield on the order of 1000 cells per 100 nanoliter well. Typically, an entire chip or the subset of through-hole wells constituting a contiguous region of platen 10 may be populated with a single strain of cells.
  • A typical procedure assay procedure, such as may be employed in pharmaceutical research, entails the subsequent addressed introduction of a test sample including one or more analytes into the through-hole wells, with selected materials introduced into subsets of through-holes that may include one or more through-holes. The test sample addressably introduced into the subsets of through-holes may contain drug candidates or known drugs. The test sample may be comprised of multiple components, introduced at the same time or sequentially. Components of the test sample may include analytes, antagonists, reagents, solvents, or any other materials and may be introduced in liquid form or otherwise. In accordance with a preferred embodiment of the invention, test samples are introduced into the through-hole wells in liquid form in order to facilitate rapid reaction via diffusion with first sample 18 already resident in liquid form in the through-holes.
  • The set of substances from which the second sample addressed to a particular through-hole site is drawn is referred to in this description and in the appended claims as a “library” of substances. In typical applications, the library is of a substantial size and thus advantageously utilizes the capability of the present invention to facilitate parallel reaction and analysis of large numbers of substances. In pharmaceutical applications in particular, libraries may be composed of between 103 and 109 substances and combinations of substances.
  • Referring now to FIG. 1 c, the throughput of a screening system incorporating arrays of through-hole sample containers may be further increased by using a continuous sheet 300 of through-holes 12. The interiors of the through-holes may be hydrophilic, as described above in reference to FIG. 1 b, and the surface of sheet 300 may be hydrophobic, as also described. Through-holes 12 may be filled in a continuous manner by passing sheet 300 through an aqueous medium 302 contained in fluid trough 304. After through-holes 12 have been filled, sheet 300 may be wound onto a spool 306 or cassette for storage followed by assay, or, alternatively, may be assayed directly. Assays may be by optical readout such as fluorescence absorbance or chemi-luminescence measurements, all of which may be performed by passing the sheet across an optical detector such as a CCD array.
  • Sample array sheets 300 are preferably produced with registered holes, either by precision production processes or in matched sets. The sheet composition may be a polymer, elastomer, or metal, including an amorphous metal. Multiple sheets may be mated in the same way that platens of through-holes may be stacked, as described below, for example, with reference to FIGS. 16-19, in order to initiate reactions, or for other purposes. FIG. 1 c shows a second sheet 308 of through-holes 310 being brought into contact with sheet 300 in mixing area 312 for mixing of the liquid contents of the respective through-holes. Examples of applications involving this embodiment include, screening genetic libraries or screening combinatorial chemical libraries contained on polymer beads. These embodiments of the invention may advantageously include extremely high throughput, reduction or elimination of high-cost automation components, and the small size of a screening system with sample handling and detection modules. The holes in the sheet may be, if desired, produced online with an array of punches or UV lasers.
  • As an example of an application in which a genetic library is screened for improved enzymes using a one step assay, an E. coli genetic library is prepared containing mutations in the beta galactosidase enzyme. The E. coli cells are grown to a density in phosphate-limited media such that there is an average of 1 cell for every 200 nl of liquid. The media also contains, MUG, a fluorogenic substrate for beta-galactosidase. A through-hole sheet is prepared with a hydrophobic exterior and hydrophilic through-holes at a density of 10̂7 per square meter. Registration holes are includes in the tape to aid in precise dispensing. Each through-hole holds 70 nl of fluid. A spool of the through-hole sheet is unwound and guided through a trough containing the cell solution, so that each through hole is filled. The sheet is then wound onto spacers and into a receiving spool. The spacers prevent smearing of the liquid and provide for gas transfer in and out of the spool. The spool is incubated in a humidified enviroment at 37° C. for 24 hours. The spool is then unwound as it is passed between a uniform photo-illumination source with a wavelength of 350 nm and a CCD imaging system with a 450 nm filter. The position in the sheet of colonies with exceptionally high enzyme activity is recorded and those colonies are retrieved using a robotic microfluid handling system for further analysis. This assay can also be performed by co-registering and mating a second, identical, sheet containing the fluorogenic substrate with the first sheet containing the bacteria. An absorbance measurement may also be performed to normalize the signal output for the number of bacteria.
  • In accordance with another alternate embodiment of the present invention, described with reference to FIG. 3, through-holes 12 may be disposed in an array within a circular sample wafer 320 having a central hole 322 for purposes of centering with respect to handling equipment.
  • Referring now to FIG. 5, platen 10 and through-holes 12 may assume other geometries than have heretofore been described, in accordance with alternate embodiments of the invention. The volumes of the through-holes may be varied as a known function of spatial location in the array. The volume of a through-hole, V=ks2l, is a function of lateral hole dimension, s, length, l and constant k that depends on the specific cross-sectional hole geometry. The cross section may be varied, as depicted in FIG. 5 a, as a known function of spatial location within the array by changing the hole's lateral dimensions as a function of position within the array. The hole volume scales as s2; thus increasing the hole dimensions by a factor of 3.3 increases the volume by a factor of 10. One example is in a plane parallel plate, the hole dimensions are increased as a linear function in one or both lateral directions.
  • Another method to produce different through-hole volumes as a function of array position is to change the distance between top and bottom surfaces of the plate defining the hole length 52, as shown in FIG. 5 b. One particular example is to incline the plate top 14 and bottom 16 surfaces at an angle to each other as in a wedge or prism, as shown in FIG. 5 c. In this example, the volume change is linear with distance along the array in the direction of surface inclination while in the orthogonal direction along a row of holes, the hole length, and thus volume, is constant. In accordance with another alternate embodiment of the invention, an array can be fabricated from a contiguous series of planar surfaces inclined to each other in either one- or two-dimensions such that the through-hole volume is different in both directions along the array. In yet a further embodiment, an array is fabricated from a non-planar surface 54 such as a surface of hemispherical curvature as shown in FIG. 5 d.
  • For each array geometry, a second array fabricated having surfaces that are the complement of the first array's geometry may be used, in accordance with alternate embodiments of the invention, to facilitates vertical stacking of the arrays, as shown in FIG. 6. Alignment of through-holes 64 of plate 60 with through-holes 66 of plate 62 is provided by inserting protruding segments 68 into corresponding indentations 70. Other array geometries with greater than four-fold rotation symmetry with respect to the array surface normal will facilitate interlocked and self-aligned stacking of arrays with matching positive and negative geometries, within the scope of the present invention. This geometrical arrangement may advantageously obviate the need for alignment pins which are typically required for registration of stacked planar arrays.
  • Applications of Arrays of Non-Uniform Volume
  • One application of arrays having through-holes of non-uniform volume is mixing of different volumes of liquids such as in a dilution sequence, as described below. An advantage of this method is that stacking of multiple arrays, as also described below, may be obviated while a wide range of dilution may be achieved.
  • Another application of arrays of either constant or non-uniform through-hole volumes is fraction collection from chromatographic elution. The through-hole array advantageously provides the ability to collect a large number of small volume fractions, which can then be further separated within the through-hole array, as described below. This, in turn, advantageously increases the resolution of chromatographic separation over prior technology.
  • Yet another application of non-standard array geometries is for massively parallel liquid chromatography or electrophoresis. More particularly, referring to FIG. 5 e, a through-hole plate 56 is employed whose thickness increases in one lateral direction (such that the plate has the shape of a prism) resulting in a linear increase in hole length with array position. A second through-hole prism 57 is brought together with the first prism 56 along their common hypotenuse 58 thereby producing an array of through-holes 12 of substantially equal length. To use this structure as a liquid chromatography column, each hole is filled with a porous gel 55 characteristic of liquid chromatography. The sample 51 to be analyzed is applied to one end of the array, as from a sample plate 53, for example, and a pressure is applied to drive fluid sample 51 through each hole. Each component in the mixture will travel at a different velocity through the gel matrix resulting in a separation of the mixture along the column length. For example, small molecules will move rapidly through the channel to its opposite end, whereas, after the same duration of time, larger molecules may typically have traveled only a fraction of that distance. The intersection of each through-hole with the hypotenuse along which the prisms are joined corresponds to different travel lengths along the chromatographic column. Separation of the array into its constituent prisms, as depicted in FIG. 5 f, gives a “snapshot” of the mixture components distributed as a function of array position. Rather than a function of time, the chromatograph is transformed into a function of position by running identical samples in an array of columns with a known inter-column delay, expressed either as a time, or, equivalently, as a length. By sequentially analyzing the material output from each through-hole along the direction of increasing plate thickness, an equivalent chromatograph can be reconstructed. Further mixture separation is possible if the gel porosity is made different in the array direction orthogonal to the wedge orientation. Decreasing gel porosity further increases the retention time for each component, thus leading to finer resolution of the mixture components. Electrophoretic separation may be achieved in an analogous manner, where an applied electric field is the driving force.
  • Fabrication of the Through-Hole Array
  • Referring now to FIG. 7, through-holes 12 may be formed in platen 10 by any of a variety of means appropriate to the material of platen 10. Through-hole forming methods include, by way of example, laser ablation by means of an ultraviolet (UV) excimer laser which may form 100 pm through-holes in glasses and polymers. Additional through-hole forming techniques include mechanical drilling, electrochemical methods such as micro-electrical spark discharge machining (EDM), employing radio-frequency pulses for ionizing removal of conductive materials, or, in accordance with other embodiments of the invention, by selective chemical or charged-particle etching techniques. Additionally, microcapillary bundles of glass fibers of varying compositions may be drawn from preform and sliced to form platens, and then selectively etched to form through-holes.
  • As shown in FIG. 7, through-holes 12 may be formed, in materials such as thermoplastics or polycarbide, by punching platen blank 400 using punch array 402 and die array 404, in conjunction with a high-pressure ram. Punching pins 406 may be formed in punch block 408 using microwire EDM, as described in detain in the following discussion, or by microetching techniques such as chemical or charged-particle etching. Additionally, punch array 402 may be formed using microsawing techniques. Die array 404 is similarly formed using microfabrication techniques known in the manufacturing arts.
  • High-density arrays of through-holes may be formed in conducting materials, including conductive (>10Ω−1-cm−1) silicon wafers, using a combination of wire and die sink EDM. EDM is typically used in the preparation of tooling dies for injection molding. As is well known, the machining process involves ionizing away the surface of a conducting material, typically a metal. EDM can be performed using a tip-based electrode, or a “wire.” Wire-EDM is used when fine surface finishes are required, or when subtle machining features not achievable with a tip electrode are desired. Traditional wire-EDM machining utilizes wire approximately 250 μm in diameter. Due to the small dimensions and high packing densities of the invention an adapted microwire EMD process is used that employs wires with diameters down to 30 μm. This system may provide surface finishes down to 100 nm, essentially a mirror finish.
  • Referring to FIG. 8, a typical method for preparing a through-hole array using microEDM machining follows a two-step process. The first step is the creation of a positive or master die. The second step uses another EDM machine called a “sink-EDM.” The sink-EDM machine uses the master as an electrode and thereby creates a negative copy in the machined conducting material. This negative copy is the resulting microarray. The master electrode can produce multiple negatives before needing to be replaced, thereby increasing manufacturing throughput for production of the chips.
  • In contrast with deep reactive ion etching—the process commonly used to produce high aspect ratio structures in silicon—the EDM technique described herein may advantageously reduce fabrication time and cost.
  • After the master die is fabricated with wire EDM, it may typically be used to sink an array of microchannels through a 0.5-mm thick silicon wafer in less than two minutes. Additional machining time, during which the master die moves largely in the plane of the array, is typically needed to enlarge the holes to the desired dimension and to improve the surface finish. The fabrication process described has been used to sink microchannels of sizes up to 10 000 elements with cross-sections <300 μm×300 μm and element spacings <500 μm. Another advantage of this technique is that it may also be used to a manufacture stainless steel precision alignment jig that is used to align the chips for mixing and optical readout, as described herein.
  • Combined with the precision alignment jig, this EDM process results in <10 μm center-to-center total error in channel spacing across the entire array. This guarantees accurate hole alignment across arrays, and when combined with the hydrophobic exterior coating, minimizes cross talk between microchannels.
  • Referring again to FIG. 7, the master die 408 produced from the process outlined above can be used to create the through-hole arrays from a plastic blank 400. In this approach the die is used as a type of punch to create the through-hole array from a solid piece of plastic. Unlike a punch, however, the master does not force its way through the plastic. Instead, by having the appropriate kinetic energy as it impacts the plastic it essentially vaporizes the solid plastic into low molecular weight gases (with some residual energy dissipated as heat). In this manner the master can be removed from the resulting through-holes without having melted into it. A slight taper on the protrusions of the master that forms the channels facilitates removal of the master from the chip. This process is similar to that which is used in the manufacturing of DVD's, except the manufacture of microwell arrays typically requires significantly greater penetration depths (up to 1 mm deep). Alternatively, plastic microwell plates may be manufactured by injection molding of metal masters formed by EDM.
  • As discussed above, with reference to FIG. 1 b, it is desirable to prevent cross-communication between the various through-holes during loading and other operations by coating the surfaces of the platen with a hydrophobic coating 22. It is also desirable to coat the inner surfaces of the through-holes with a hydrophilic coating 24 so that they retain fluids. In accordance with embodiment s of the invention, the inner coating 24 may be chemically blocked to prevent non-specific binding or derivatized with affinity ligands.
  • In accordance with preferred embodiments of the invention, a dense array 10 of through-holes 12 is produced in silicon and coated in silicon oxide by oxidation. The surfaces of the conductive silicon are covered in a thin oxide layer. The wafer is then cleaned by soaking in a mixture of hydrogen peroxide and sulfuric acid, or other caustic solvent cleaning solution, to remove organic materials. Clean silicon oxide thus produced has a high surface energy. The top and bottom faces of the arrays are made hydrophobic by exposing them to vapor from a solution containing an appropriate silanizing agent (such as polydimethylsiloxane, sold as Glassclad 216™ by United Chemical Technologies, Inc.) in a volatile solvent. The silanizing agent reacts with the hydroxyl and silanol groups on the array surface and creates covalent bonds to hydrophobic alkyl groups.
  • The resulting coated arrays can be uniformly loaded with aqueous solutions by simply dipping the array into it. The liquid instantaneously fills the channels through capillary pressure, but does not wet the other surfaces. Hydrophobic coatings produced in this way are stable under high humidity and they can be used repeatedly over several days. Other surface chemistries may be exploited to attach hydrophobic chemical groups to the faces of arrays made from other materials. For example a gold-coated surface reacts with alkane thiols to attach a hydrophobic alkyl groups, as discussed by C. D. Bain et al., J. Am. Chem. Soc., vol. 111, pp. 321-325 (1989), which reference is incorporated herein by reference.
  • In accordance with other embodiments of the present invention, the surface chemistry of the array faces may also be selectively modified by substituting other silanizing agents for the polydimethylsiloxane telomer. This method may advantageously prevent aqueous solutions from adhering to array faces during array loading and also act as physical barriers between the aqueous solutions in adjacent through-holes. During this process a positive pressure of inert gas is applied to the opposite side of the array. The positive pressure within the through-holes prevents the silanizing vapor from reaching the interior surfaces. This method advantageously allows removal and reaplication of the hydrophobic coating.
  • Array Loading Techniques Dip Loading
  • Referring now to FIGS. 9 a-9 c, array 10 of through-holes 12 may be filled using techniques either that directly address particular through-holes, or techniques that fill the entirety of the array according to a specified pattern based on composition, concentration of a substance, etc.
  • Dip loading may be employed, for example, in order to fill an entire array with the same solution. A through-hole array 10 is produced and chemically treated to make the array faces hydrophobic and the through-hole surfaces hydrophilic. Plate 10 with through-holes 12 is first lowered into a container 90 containing a first liquid 82, as shown in FIG. 9 a. Once plate 10 is fully immersed in the first liquid, it is shaken so that first liquid 82 displaces the air in each of the through-holes, and all the through-holes 12 are filled with first liquid 82. All means of replacing the air in the through-holes is within the scope of the present invention, whether by shaking, applying a vacuum to one side of plate 10, employing electrostatic forces, tilting the plate so that the air is displaced by virtue of its buoyancy, all cited as examples and without limitation, or else by other means. After through-holes 12 have been filled, plate 10 is withdrawn from first liquid 82, as shown in FIG. 9 c. The array is typically withdrawn slowly such that the surface of the fluid in the reservoir pulls excess liquid off of the non-wetting array surface. Alternatively the array may be filled with water by spraying the array with water or by sonicating the array in a reservoir of water in order to remove trapped air bubble prior to dip loading. In this case, less agitation is required to uniformly fill the array with solution.
  • Both convection and diffusion can be used to induce mixing between fluids in microchannel arrays. This can be demonstrated by filling a microarray with aqueous solution of blue dye and by submerging the microarray into a beaker of still water to induce dilution via diffusion. A small mechanical disturbance (such as a tap to the side of the beaker) causes rapid replacement of the blue dye by water. Alternative methods for filling the array with water include spraying the array with water or sonicating the array in a reservoir of water in order to remove trapped air bubble prior to dip loading are also within the scope of the present invention.
  • Similarly, an array having hydrophilic faces and hydrophobic through-holes may be uniformly filled with a low surface tension liquid such as an alkane.
  • Loading by Dragging Droplet Along Array Surface
  • Droplet dragging is performed to load an entire array or a group of channels within a single array with the same fluid. It is appropriate for loading samples which must be conserved because of cost or availability. To perform a loading operation by dragging, a droplet of the loading solution is formed on the end of a syringe needle, micropipette or other fluid dispensing capillary. The drop is placed on the array face over the through-holes to be filled. The capillary is then moved to drag the drop across the surface of the array and over the channels to be filled. Surface tension maintains contact between the capillary tip and the fluid drop. When the drop is depleted additional fluid is dispensed until all desired through-holes have been filled. The capillary and any remaining fluid in the drop are then withdrawn from the array surface.
  • Transfer of Liquids Between Microtiter Plates to Through-Hole Array
  • In accordance with further embodiments of the invention, a method is provided to transfer liquids from standard microtiter plate formats (96-, 384-, or 1536-wells, for example) to a single array of through-holes or multiple, vertically stacked arrays in which holes in the same spatial location from one array to the next are co-registered. As used herein, the term “registration” refers to aligning a through-hole plate with at least one other through-hole plate, such that the tops of a plurality of through-holes of one of the plates coincide with the bottoms of a plurality of corresponding through-holes of the other plate or plates, thereby creating a plurality of contiguous channels.
  • All of the filling examples given can be performed in parallel on a single array stack with multiple capillary tubing arrays filled with liquids from different microtiter plates or the same microtiter plate. Fluid may be transferred from chemical libraries stored in 96- and 384-well microtiter plates quite rapidly with these methods. For example, if 100 arrays with 105 through-holes per array are to be filled with liquid stored in 96 well plates, and if each transfer operation (plate exchange, fluid loading and transfer) takes approximately 20 s, then the requisite arrays may be filled within approximately six hours. The filling operations take place preferably in an environmentally-controlled chamber (temperature, humidity, atmosphere, etc.), as described below.
  • Transfer of Liquids from Microtiter Plates to One or More Through-Hole Array
  • Referring now to FIGS. 10 and 11, further embodiments of the invention provide an apparatus and methods that are particularly suited to the transfer of liquids from standard microtiter plate formats (such as 96-, 384-, or 1,536-well formats) to a single array of through-holes or multiple, vertically stacked arrays in which holes in the same spatial location from one array to the next are co-registered. Producing such stacks of arrays may be employed advantageously for producing replicates of molecular or cellular libraries.
  • Capillary Tube Array
  • Viewed in cross-section, a capillary tube array 100 is constructed from capillary tubing 102 with an external diameter that fits precisely into the through-holes of a through-hole microwell array 10. Tubing array 100 is designed such that tubing 102 at one end 104 has a center-to-center spacing 106 equal to the spacing between holes in through-hole array 10 (or, alternatively, to an integral multiple of the inter-hole spacing) and tubing at the opposite end 108 has a center-to-center spacing equal to the center-to-center spacing 110 of wells 112 in a microtiter plate 114. Plates 116, 118, and 120, with through-holes having these and intermediate separations serve as jigs to hold the tubing in a regular array. Additional through- hole plates 116, 118, and 120 placed between the two ends may advantageously serve as spacer jigs providing additional support for the tubing array as the center-to-center spacing varies over the tubing length.
  • In addition to filling a single through-hole microwell array 10, the technique depicted in FIG. 10 may be advantageously employed for filling an entire stack 122 of hole-registered microwell arrays. The internal volume of each tube 102 in the capillary array 100 is slightly greater than the total volume of a column of aligned holes in the array stack 122. For example, if the through-hole dimensions in the array are 250 μm×1000 μm, giving a volume per through-hole equal to 62.5 nl, then the volume of one set of holes in a stack of 100 arrays is 6.25 μl (100×62.5 nl). Capillary tubing with an internal diameter of 200 μm and an external diameter of 245 μm is readily available; thus a minimum tube length of 200 mm stores the volume of fluid needed to fill this set of through-holes.
  • One end 108 of tubing array 100 is inserted into the wells of a microliter plate 114, each tube being inserted into a corresponding well 112. Next, a negative pressure difference is applied across the length of tubing 102 to draw liquid, in direction 126, from each well into its corresponding tube. Negative pressure could be applied to each tube individually, or as shown in FIG. 10, the ends of the tube array can terminate in a chamber 124 that can be partially evacuated. After filing each tube of the array, the microtiter plate 114 is removed. The liquid can be stored in the tubing array for an indefinite period of time, either frozen or in a humidified environment. It can also be readily transported to another location in this format. Multiple tubing arrays can be filled from the same microtiter plate (assuming there is sufficient volume of liquid per well) or different tubing arrays can be filled from different microtiter plates.
  • Proximity Filling
  • Embodiments of the invention also provide for methods for filling a stack of arrays, as shown in cross-section in FIG. 11, by bringing the end of a tubing array 130 into close proximity with a matching set of through-holes in the array stack 122. The tubing array can be aligned relative to the array stack by an alignment plate 128 with through-holes having the same center-to-center spacing as the through-holes into which fluid is placed. Each array 10 in the stack 122 is spaced a small distance s that may be, but is not limited to, an equal distance to the through-hole spacing. Application of pressure to the end of the tubing array, placed inside a pressurized container 132, forces fluid from each capillary tube 102 into the opposing through-hole. After the through-hole is filled, a liquid drop can begin to grow in the space between the two plates. When the drop reaches a size that it contacts the through-hole in the plate above it, surface tension draws some fluid into the through-hole. Once the fluidic bridge is established, liquid can flow into the through-hole, driven by the constant pressure applied to the opposite end of the tubing array. With no applied pressure, the drop retreats into the through-hole, the fluidic bridge between each plate is broken, and the separation of array plates after filling can be facilitated (i.e., because there is generally no surface tension that needs to be overcome). Successive filled plates 10 are then withdrawn, and the tube array may be retracted in direction 128. Each vertically registered set of through-holes may thus act as a channel for fluid flow. The hydrophobic coatings on the exterior surface of the arrays prevent liquid from flowing into adjacent holes. This technique can also advantageously be used to create replica plates of a cell library by applying a cell suspension with a pressure uniformly to the array stack.
  • Inter-Hole Spacing Matched to a Microtiter Plate
  • A method for filling a through-hole array stack, in accordance with alternate embodiments of the invention, is shown in FIG. 12. This method uses a through-hole array 10 having the same lateral dimensions as a microtiter plate 114 and having a hole spacing that is an integral fraction of the well spacing in the microtiter plate 114. When the array is placed on top of the plate, one or more through-holes 12 align with respect to each well 112 in microtiter plate 114. An array 130 of syringes 132 with a center-to-center spacing equal to the well spacing can thus be positioned over a stack of through-holes registered with respect to each other and the microtiter plate. The syringe array is inserted through a through-hole plate 134, such that the plate is a mechanical guide for the syringe tubing as it is moved relative to the array stack. The syringe plungers are mechanically coupled and actuated by a mechanical or electromechanical driver module 136, such that liquid is drawn into or expelled from each syringe in parallel. The syringe tubing outside diameter is preferably sized relative to the through-hole lateral dimensions to give a sliding fit, and the tubing can have a length suitable to allow insertion through the array stack 122 and into the liquid contained in the microtiter plate wells 112.
  • The volume of liquid withdrawn into each syringe preferably equals the volume of liquid in a column of aligned through-holes in the array stack. The liquid can then be dispensed as the syringe array is retracted from the array stack, and the rate of dispensing can be synchronized with the rate of withdrawal, such that each through-hole addressed by the syringe array is filled. Once this operation is completed, either a new set of holes in the array stack can be filled from the same microtiter plate or the syringe array can be washed and different set of holes filled from a different microtiter plate.
  • All of the filling examples given above may be performed in parallel on a single array stack with multiple capillary tubing arrays filled with liquids from different microtiter plates or the same microtiter plate. The time to transfer fluid from chemical libraries stored in 96 and 384 well microtiter plates with these methods can be quite rapid. Assuming, for example, that 100 arrays with 10,000 through-holes per array are to be filled with liquid stored in 96 well plates, and that each transfer operation (plate exchange, fluid loading and transfer) takes approximately 20 seconds, then it would take approximately six hours to fill all of the arrays. The filling operations can take place in an environmental controlled chamber (temperature, humidity, atmosphere, etc.). The invention also provides a method for screening compound libraries to predict the ability of each compound to be absorbed by a patient.
  • Transfer from a Microtiter Plate with an Array of Flexible Members
  • As now described with reference to FIGS. 12 b and 12 c, fluid can be transferred from individual wells 112 of a microtiter plate 114 with an array 140 of flexible members 142, e.g., shape memory alloy fibers. The fiber diameter is equal to or less than the inside dimension of the through-holes 12 in the array 10 into which fluid will be transferred. The number of fibers in the bundle may, for example, be equal to the number of wells in the microtiter plate 114. The ends of the fibers at one end of the bundle can have a center-to-center spacing equal to the spacing of the holes in the through-hole array, while the ends of the fibers at the opposite end can have a center-to-center spacing equal to the spacing of wells in the microtiter plate. The fibers can be held in place with a series of through-hole jigs designed to increase the spacing between fibers from one end of the bundle to another. Once fixed in place, shape memory alloy fibers can be heated above their critical transition temperature to make the imposed fiber curvature permanent. After they are cooled to room temperature, the fibers can be removed from the holding jig, with the change in fiber center-to-center spacing intact. The close packed end of the fiber bundle can then be inserted into the through-hole array into which fluid from each well in the plate is to be placed. The opposite end can be arranged such that each fiber is positioned above a well in the microtiter plate, and the ends of the fibers can be immersed in the fluid contained in each well. On retraction of the fiber bundle from the microtiter plate, as shown in FIG. 12 c, a small volume drop 144 remains attached (e.g., by surface tension) to the end of each fiber 142. A force may be applied to the opposite end of the fiber bundle to pull the bundle through the holes 12 of the through-hole array 10, such that the fluid is brought into contact with the corresponding through-holes. As the fibers 142 are pulled through the hole 12, surface tension acts to hold the liquid in the through-hole as the fiber is removed.
  • Successive Dilution
  • Addressable loading may also be employed to fill a series of through-holes with different and specified concentrations of the same solute. A chosen series of through-holes is filled with a quantity of solvent, denoted Z nanoliters (nL), either by dipping the holes into the solvent or by dispensing the solvent from a microsyringe 232 (shown in FIG. 21), or other fluid transfer device. This is shown as the first step 120 in the flow chart of FIG. 13. In step 121, microsyringe 232, or another fluid transfer device, is filled with an X molar solution of the solute, and is then positioned over the first through-hole. Y nL of this solution, where (Y+Z) nL is sufficient volume to overfill the hole and to create positive menisci, is expelled such that it forms a droplet at the end of the syringe tip. The syringe tip is lowered until the solvent droplet contacts the surface of the solution, causing the two liquids to mix 122 and produce a solution of concentration YX/(Y+Z) molar. The outer surface of the syringe tip and the faces of the array must be nonwetting toward the solution being dispensed. The syringe plunger is then withdrawn in step 123 to suck up Y nL of the diluted solution. The syringe tip is positioned above the next (N+1) through-hole 124 and Y nL of the diluted solution is dispensed into the solvent to dilute 125 by another factor of Y/(Y+Z). The process is repeated so as to dispense solution into a series of individual through-holes, each time diluting by Y/(Y+Z).
  • Chemical Gradient Methods
  • In accordance with preferred embodiments of the invention, a particular chemical species is not loaded uniformly into all the holes of an array, but, rather, a gradient of chemical species is created in at least one dimension in a two-dimensional through-hole array. As used in this description and in any of the appended claims, the term “gradient” refers to its ordinary mathematical meaning, i.e., a variation, along one or more directions, of a specified quantity. The directions, here, are taken along the surface of an array of through-holes. Thus, a gradient in the concentration of a specified chemical species in the through-holes may be said to exist with respect to a particular direction, or, for that matter, with respect to various directions. Thus, a particular gradient may be, but need not be, monotonic, and the concentration of a species, in a specified direction, may rise and fall.
  • Referring now to FIGS. 9 a-9 c and 14 a-14 c, dip loading methods described above may be used for creating a concentration gradient in an array of through-holes. In accordance with such methods, the array is dipped into a chemical solution at a controlled rate such that the chemicals in the solution have different amounts of time to mix with, or react with, substances by diffusion into various through-holes.
  • After an array has been filled, by dip loading, with first liquid 82, as discussed above with reference to FIGS. 9 a-9 c, plate 10 is subsequently lowered into a second container 84 containing a second liquid 86, as shown in FIG. 14 a. Since the through-holes of plate 10 are now filled with the first liquid 84, the second liquid 86 may diffuse into, mix with, displace, or otherwise react with, the contents of through-holes 12. Each of these processes is characterized by a rate. Instead of fully immersing the plate until the particular reaction, physical or chemical, runs to completion, plate 10 may be partially immersed into second liquid 86, as shown in FIG. 14 b, for a specified duration of time, and then withdrawn, wholly or partially, as shown in FIG. 14 c. If plate 10 is reimmersed in liquid 86, to a different depth 88 and for a newly specified duration of time, certain of the through-holes 12 will contain material subject to differential physical or chemical processes. Thus, a gradient will have been created with respect to the direction z along the array of through-holes. Naturally, this process may be repeated or otherwise modified to create a specified gradient.
  • In accordance with alternate embodiments of the invention, other means may be employed in order to spatially modulate the concentration of a specified species allowed to diffuse into a through-hole located at a particular position within the array. Referring to FIG. 14 d, a side view is shown of through-hole array plate 10 immersed in liquid 86. Diffusion of liquid 86 into through-holes 12 is modulated by membrane or filter 87, for example, that, by virtue of its tapered shape or otherwise, gives rise to a gradient of a specified species with respect to the position of a through-hole within the array.
  • In a related example, by slowly lowering a through-hole array filled with a gel forming solution thin-edge first into a polymerization initiating mixture different gel densities may be obtained within in holes. By rotating plate 10 by 90° about axis y transverse to the surface of the plate, and then slowly dipping into a reagent that derivatizes the gel with a free cationic moiety, and then rotating another 180° about y and slowly dipping into a reagent that derivatizes the gel with an anionic moiety, a two-dimensional size/charge selection matrix is created that is useful for separating protein mixtures in a manner similar to a 2-D polyacrylamide gel, but with the advantage of greater separation speed and greater access to the separated proteins.
  • In another embodiment, concentration gradients within through-hole arrays are use to optimize the concentrations of A and B in a two-component reaction between Reagent A and Reagent B. The mixing process is described in greater detail below. Referring to FIG. 15, a first through-hole array, shown in . cross section and designated generally by numeral 90, is loaded with Reagent A using a chemical gradient method that results in the through-holes along one row, in the x direction, of the array will have the same concentration of A and the concentration gradient is along the array columns, in the z direction. A second array, designated generally by numeral 92, is loaded with Reagent B such that the concentration gradient is along the rows (x direction) of the array and through-holes along a column (z direction) of the array have all the same concentration. Bringing the two arrays in contact causes mixing between aligned through-holes 12. Along each row or column of the combined arrays the concentration of A relative to B in the mixture changes in a regular and known manner. Along the array diagonal, the concentrations of A and B in the mixed liquids are equal and but changing in overall concentration.
  • Variants from the basic scheme for creating a known two-dimensional gradient in the relative proportions of A and B include loading the A and B arrays by dipping along a diagonal, by the orthogonal combination of two arrays each with a 2-D gradient of A relative to B or by dilution with arrays loaded with Reagent A or B where some of the through-holes are empty. Analysis of the through-hole contents readily determines optimal reaction conditions since the reaction conditions in each through-hole are known. Gradients created in the solution with respect to quantities other than solute concentration are also to be understood to be within the scope of the present invention, and may include, without limitation, such characteristics as temperature, electric field, magnetic field, etc.
  • Mixing and Dilution
  • As discussed in greater detail in the following sections, microwell plates may advantageously be stacked for such purposes as mixing or dilution. One such application is the apportionment among through-holes of a sample of cells. Considering, for purposes of example only, through-holes on a single plate that are 250 μm square and 500 μm deep. When three such microchannel plates are stacked (as would be the case in a 2-step assay), the total volume of a single channel (i.e., the combined volume of three through-holes) is ˜100 nL. If the entire channel is filled from a dense yeast cell culture (˜107/mL) each channel then contains approximately 103 yeast cells. Based upon a yeast cell volume of 70 μm3, the maximum number of cells per 100 nL channel is on the order of 106, consonant with a typical minimum of 100 cells per microchannel is required to compensate for variable yeast cell response to the bioassays.
  • Referring to FIGS. 15 -19, mixing and dilution are shown between the contents of two substantially planar through-holes array plates 90, 92 (i.e., array plates having neither flanges nor indentations). In FIG. 15, in particular, a cross-sectional view is shown of portions of two through- hole arrays 90, 92. Both the top and bottom surfaces of each platen are hydrophobic, as discussed in detail above. The through-hole walls 24 are preferably hydrophilic. Through-holes 12 of both platens are overfilled with high surface energy fluids 150, such as aqueous solutions, for example, such that each through-hole 12 has a positively-curved meniscus 152 protruding above the platen surface 154.
  • Alternatively, the platen surfaces 154 may be hydrophilic and a sufficient amount of a low surface energy fluid, such as an alkane, for example, is loaded into the through-holes 12 to form positive menisci 152.
  • FIG. 16 shows a cross-sectional view of the two platens 90, 92 after the surfaces of the liquids 150 in co-registered through-holes 12 are brought into contact. The release of the surface tension drives convective mixing between fluids contained in the opposite platens 90, 92. Once the two surfaces of the platens themselves have been brought into contact, each set of two-coregistered through-holes forms a longer channel 170, as shown in FIG. 17. Mixing proceeds within the longer channels by a combination of convection and statistical diffusion.
  • Referring to FIG. 18, in cases where through-holes 12 of one or both of arrays 90, 92 are underfilled (such that there is an air-gap separating the fluid 180, 182 when the plates are contacted), the application of slight positive pressure, designated by arrows 184, may be employed to bring the surfaces of the two fluids into contact while a small gap 188 is still present between the surfaces of the two platens 90, 92.
  • In yet other embodiments of the invention, with reference to FIG. 19, first platen 90 is filled with sufficient fluid 150 to form positive menisci 190, 192, while a second platen 92 is filled with an amount of fluid 194 that is insufficient to form positive menisci, but sufficient to make contact with the surface 190 of the menisci of the first platen when the two platens are stacked. Mixing proceeds by bringing the liquid surfaces into contact as previously described.
  • Control of Environmental Factors
  • In accordance with embodiments of the invention, methods and apparatus are provided for maintaining high relative humidity levels (typically above 95%) in the environment surrounding through-hole arrays during various operations of the invention. Maintenance of high humidity levels may advantageously minimize, if not eliminate, evaporation of solutions contained in the through-hole arrays. The level of humidity required to keep the volume loss at an acceptable level depends upon the length of time needed to perform the various desired operations, as well as on such factors as the ambient temperature, and the volume of fluid contained in each through-hole. At a temperature of 21° C., the evaporative loss expected from a droplet of water may be predicted in accordance with Fick's law, as depicted in FIG. 20. The time, in seconds, for 10% evaporative loss is plotted for a 50 μL droplet (by curve 200) and for a 50 nL droplet (curve 202) as a function of relative humidity. Unless humidity levels are close to 100%, the smaller droplet will evaporate very rapidly, with a 10% mass loss in 11 seconds at 65% relative humidity.
  • Humidity Control During Addressable Loading and Unloading
  • With reference to FIG. 21, and apparatus is provided for advantageously reducing the volume of the enclosed environment subject to humidity control. In accordance with embodiments of the invention, a through-hole array 10 is mounted above a reservoir 210 of water. A 96-well, 384-well or higher density microtiter plate 212 is mounted above the same reservoir proximal to the through-hole array. Reservoir 210 may contain a water absorbent material such as a sponge 214 to keep the water from sloshing when the reservoir is moved. An optically transparent plate 216 is placed on top of the reservoir separated from the walls of the reservoir by a thin layer 218 of a viscous non-hydroscopic fluid such as silicone grease, for example. One or more small holes 220 are drilled through transparent plate 216. Through each hole a microsyringe needle 222, microcapillary, pin, cannula, or other fluid transfer element is inserted.
  • Computer control of motorized translation stages may be provided, such as along axis y to move the fluid transfer element 224 towards and away from the top face 226 of the through-hole array 10. Fluid transfer element 224 may include syringe 232 with plunger 234 fluid transfer element 224, for example. Additionally, the arrays and reservoir may be moved in the plane of the x and y axes, preferably by motorized stage 228 under control of controller 230, with respect to the fluid transfer element. If indicated, further computer-controlled stages may be provided, for example, to actuate the fluid transfer device. Transfer of materials between the microtiter plate and through-hole array proceed by means described above in reference to FIGS. 10-12.
  • Additional reservoir(s) containing solution(s) for cleaning/sterilizing the fluid transfer element may also be provided within the confines of chamber 236 which encloses the apparatus heretofore described. Alternatively the water in the large reservoir 210 may be used for this purpose. An illumination source 238 may also be provided for to illuminate the array for optical inspection during loading/unloading, either visually or by means of a video camera 240.
  • Humidity Control During Dip Loading and Mixing
  • An environment enclosure 236, as shown in FIG. 21, may also be utilized in order to preventing evaporation from the through-hole arrays during dip loading and mixing. Sealable chamber 236 is large enough to contain the array and necessary apparatus for performing such desired operations as are required, including, for example, motors and translation stages 228, as well as an alignment jig 156 (shown in FIG. 15) for mixing operations.
  • Cool water vapor is generated by an external humidifier and injected into the chamber through a port. An ultrasonic humidifier is able to generate enough vapor to maintain humidity levels above 95% in a 0.13 cubic meter chamber. A circulation system consisting of a fan and baffles is provided to distribute the water vapor uniformly throughout the chamber. Various doors, hatches and iris ports may be provided for accessing the interior of the chamber. The temperature within the box may be controlled by a resistive heater and an electronic temperature controller. Such a chamber may be assembled by attaching a second humidifier to a commercial infant incubator.
  • Any surface which must be kept dry, including optical windows, and corrodible metals may be heated slightly above the ambient to prevent condensation. A humidity sensor is also provided to monitor humidity levels inside the chamber. All computer and electronic hardware are placed outside the chamber and are connected to components inside the chamber via a wire feedthrough.
  • The through-hole array may be filled within the chamber, or loaded externally and transferred into to the chamber in a humidified sealed cassette.
  • Humidity Control During Optical Analysis and Transfer Between Loading Stations
  • Referring now to FIG. 22 a, an exploded perspective view is provided of a compact portable cassette, designated generally by numeral 250, for maintaining relative humidity levels above 95% when the array is removed, for optical analysis, for example, from the humidified loading chamber 236 (shown in FIG. 21). FIG. 22 b provides a side view, in cross section, of the humidified cassette 250 of FIG. 22 a. Through-hole array 10 and an aqueous microlens array 252 are mounted in a compact, sealable enclosure. The though-hole array is positioned and held in place by means of alignment pins 256 or some other means. Both the array mount 258 and cover holder 260 are covered by an optically transparent plate 262 secured to the mount and cover by adhesive or other mechanical means. The mount may also include magnets, pins, grooves or other physical features to facilitate position the array inside optical analysis equipment.
  • The humidity is raised by passive evaporation from water contained in a transparent glass microcapillary bundle 252 (or, ‘microchannel array’) mounted beneath the through-hole array and secured by set screws or some other mechanical means. (Glass microcapillary bundles are manufactured by Schott Fiber Optics, Southbridge, Mass.). Typical dimensions for each capillary 264 are diameter of 200 microns and depth of 1 mm. The top 266 and bottom 268 of the microchannel array 252 are made hydrophobic according to the procedures described above. The arrangement of the capillaries 264 in the array need not be regular. Water 270 in the capillaries forms a set of microlenses which serve to diffuse light from light source 272 across the array and thus provide a uniform illumination field for optical analysis. The use of liquid microlens arrays, generally, is within the scope of the present invention.
  • Another advantage of the aqueous lens array is that the water is held in place by surface tension. Thus the operator need not keep the humidity cassette level, or avoid applying the typical accelerations that occur when the plate is carried around a room and placed on horizontal surfaces. Also the cassette may be agitated during incubation to promote cell growth inside the arrays.
  • Control of Other Environmental Factors
  • Pressure, light and temperature, are controlled by enclosing the array in an appropriately constructed chamber and then controlling the environment inside the chamber by conventional means. To prevent evaporation such chamber must also be equipped to maintain high relative humidity.
  • Having thus described various illustrative embodiments of the present invention and some of its advantages and optional features, it will be apparent that such embodiments are presented by way of example only and are not by way of limitation. Those skilled in the art could readily devise alternations and improvements on these embodiments, as well as additional embodiments, without departing from the spirit and scope of the invention. All such modifications are within the scope of the invention as claimed.

Claims (11)

1.-8. (canceled)
9. A system for performing a biological assay, comprising
a first spool configured to unwind a sheet comprising a plurality of containers for holding a biological solution;
a second spool configured to wind the sheet;
a dispenser configured to load the containers;
wherein the dispenser loads the containers when the sheet is unwound from the first spool.
10. The system of claim 9, further comprising a sheet comprising a plurality of containers for holding a biological solution.
11. The system of claim 10, wherein the sheet comprises a plurality of registration holes.
12. The system of claim 10, further comprising a mixing area for mixing liquid into at least some of the containers.
13. The system of claim 9, further comprising an optical detector configured to detect at least one of a fluorescence or a chemi-luminescence of a biological solution contained within at least some of the containers.
14. The system of claim 9, wherein the plurality of containers comprises an array of through-holes.
15. The system of claim 14, wherein sheet comprises a hydrophobic exterior and hydrophilic through-holes.
16. A method of performing a biological assay, comprising
providing a plurality of containers for holding a biological solution;
unwinding the sheet from a first spool;
winding the sheet onto a second spool;
loading a biological solution into the containers from a dispenser as the sheet is unwound from the first spool.
17. The method of claim 16, further comprising mixing liquid into at least some of the containers.
18. The method of claim 16, further comprising detecting at least one of a fluorescence or a chemi-luminescence of a biological solution contained within at least some of the containers.
US14/582,122 1998-01-12 2014-12-23 Systems for Filling a Sample Array by Droplet Dragging Abandoned US20170028376A9 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US14/582,122 US20170028376A9 (en) 1998-01-12 2014-12-23 Systems for Filling a Sample Array by Droplet Dragging

Applications Claiming Priority (11)

Application Number Priority Date Filing Date Title
US7117998P 1998-01-12 1998-01-12
US09/225,583 US6387331B1 (en) 1998-01-12 1999-01-05 Method and apparatus for performing microassays
US23953800P 2000-10-10 2000-10-10
US09/850,123 US6893877B2 (en) 1998-01-12 2001-05-07 Methods for screening substances in a microwell array
US10/820,679 US20040191924A1 (en) 1998-01-12 2004-04-08 Reformatted through-hole arrays
US10/958,969 US7547556B2 (en) 1998-01-12 2004-10-05 Methods for filing a sample array by droplet dragging
US12/468,417 US8029745B2 (en) 1998-01-12 2009-05-19 Systems for filling a sample array by droplet dragging
US13/229,208 US20110319300A1 (en) 1998-01-12 2011-09-09 Systems for Filling a Sample Array by Droplet Dragging
US201314059433A 2013-10-21 2013-10-21
US14/301,325 US20150126412A1 (en) 1998-01-12 2014-06-10 Systems for filling a sample array by droplet dragging
US14/582,122 US20170028376A9 (en) 1998-01-12 2014-12-23 Systems for Filling a Sample Array by Droplet Dragging

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US14/301,325 Continuation US20150126412A1 (en) 1998-01-12 2014-06-10 Systems for filling a sample array by droplet dragging

Publications (2)

Publication Number Publication Date
US20150298089A1 true US20150298089A1 (en) 2015-10-22
US20170028376A9 US20170028376A9 (en) 2017-02-02

Family

ID=22099761

Family Applications (6)

Application Number Title Priority Date Filing Date
US09/225,583 Expired - Lifetime US6387331B1 (en) 1998-01-12 1999-01-05 Method and apparatus for performing microassays
US09/710,082 Expired - Lifetime US6743633B1 (en) 1998-01-12 2000-11-10 Method for performing microassays
US10/796,856 Abandoned US20040171166A1 (en) 1998-01-12 2004-03-09 Method and apparatus for performing microassays
US12/794,353 Abandoned US20110065590A1 (en) 1998-01-12 2010-06-04 Method and Apparatus for Performing Microassays
US14/301,325 Abandoned US20150126412A1 (en) 1998-01-12 2014-06-10 Systems for filling a sample array by droplet dragging
US14/582,122 Abandoned US20170028376A9 (en) 1998-01-12 2014-12-23 Systems for Filling a Sample Array by Droplet Dragging

Family Applications Before (5)

Application Number Title Priority Date Filing Date
US09/225,583 Expired - Lifetime US6387331B1 (en) 1998-01-12 1999-01-05 Method and apparatus for performing microassays
US09/710,082 Expired - Lifetime US6743633B1 (en) 1998-01-12 2000-11-10 Method for performing microassays
US10/796,856 Abandoned US20040171166A1 (en) 1998-01-12 2004-03-09 Method and apparatus for performing microassays
US12/794,353 Abandoned US20110065590A1 (en) 1998-01-12 2010-06-04 Method and Apparatus for Performing Microassays
US14/301,325 Abandoned US20150126412A1 (en) 1998-01-12 2014-06-10 Systems for filling a sample array by droplet dragging

Country Status (9)

Country Link
US (6) US6387331B1 (en)
EP (3) EP2286918B1 (en)
JP (3) JP4271371B2 (en)
AT (2) ATE477850T1 (en)
AU (1) AU2102699A (en)
CA (1) CA2316912C (en)
DE (2) DE69942697D1 (en)
ES (1) ES2350702T3 (en)
WO (1) WO1999034920A1 (en)

Families Citing this family (167)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6327031B1 (en) * 1998-09-18 2001-12-04 Burstein Technologies, Inc. Apparatus and semi-reflective optical system for carrying out analysis of samples
US7122338B2 (en) * 1995-11-14 2006-10-17 Biocontrol Systems, Inc. Method for quantification of biological material in a sample
US6794127B1 (en) 1997-06-16 2004-09-21 Diversa Corporation Capillary array-based sample screening
US6972183B1 (en) 1997-06-16 2005-12-06 Diversa Corporation Capillary array-based enzyme screening
US6143496A (en) 1997-04-17 2000-11-07 Cytonix Corporation Method of sampling, amplifying and quantifying segment of nucleic acid, polymerase chain reaction assembly having nanoliter-sized sample chambers, and method of filling assembly
US6090251A (en) 1997-06-06 2000-07-18 Caliper Technologies, Inc. Microfabricated structures for facilitating fluid introduction into microfluidic devices
US7019827B2 (en) 1997-06-16 2006-03-28 Diversa Corporation GigaMatrix holding tray having through-hole wells
US6893877B2 (en) * 1998-01-12 2005-05-17 Massachusetts Institute Of Technology Methods for screening substances in a microwell array
ATE477850T1 (en) * 1998-01-12 2010-09-15 Massachusetts Inst Technology DEVICE FOR PERFORMING MICROTESTS
US7282240B1 (en) * 1998-04-21 2007-10-16 President And Fellows Of Harvard College Elastomeric mask and use in fabrication of devices
ATE341003T1 (en) * 1999-02-16 2006-10-15 Applera Corp DEVICE FOR HANDLING BEADS
US6296702B1 (en) 1999-03-15 2001-10-02 Pe Corporation (Ny) Apparatus and method for spotting a substrate
WO2000056456A1 (en) * 1999-03-19 2000-09-28 Genencor International, Inc. Multi-through hole testing plate for high throughput screening
US6555389B1 (en) 1999-05-11 2003-04-29 Aclara Biosciences, Inc. Sample evaporative control
US6977145B2 (en) 1999-07-28 2005-12-20 Serono Genetics Institute S.A. Method for carrying out a biochemical protocol in continuous flow in a microreactor
EP1214139A2 (en) * 1999-09-17 2002-06-19 Millipore Corporation High throughput screening card
DE19964337B4 (en) * 1999-10-01 2004-09-16 Agilent Technologies, Inc. (n.d.Ges.d.Staates Delaware), Palo Alto Microfluidic microchip with bendable suction tube
US20020151040A1 (en) 2000-02-18 2002-10-17 Matthew O' Keefe Apparatus and methods for parallel processing of microvolume liquid reactions
CA2400644C (en) * 2000-02-18 2009-07-14 Board Of Trustees Of The Leland Stanford Junior University Apparatus and methods for parallel processing of micro-volume liquid reactions
US20040014102A1 (en) * 2000-02-22 2004-01-22 Shiping Chen High density parallel printing of microarrays
CA2399189A1 (en) 2000-02-22 2001-08-30 Genospectra, Inc. Microarray fabrication techniques and apparatus
CN1404415A (en) * 2000-02-22 2003-03-19 基因谱公司 Microarray fabrication techniques and apparatus
AU2001286759A1 (en) * 2000-08-25 2002-03-04 Genospectra, Inc. Three-dimensional probe carriers
WO2002030561A2 (en) * 2000-10-10 2002-04-18 Biotrove, Inc. Apparatus for assay, synthesis and storage, and methods of manufacture, use, and manipulation thereof
US20100261159A1 (en) 2000-10-10 2010-10-14 Robert Hess Apparatus for assay, synthesis and storage, and methods of manufacture, use, and manipulation thereof
EP1410044A2 (en) * 2000-11-08 2004-04-21 Burstein Technologies, Inc. Interactive system for analyzing biological samples and processing related information and the use thereof
US20030007894A1 (en) * 2001-04-27 2003-01-09 Genoptix Methods and apparatus for use of optical forces for identification, characterization and/or sorting of particles
US6936811B2 (en) * 2000-11-13 2005-08-30 Genoptix, Inc. Method for separating micro-particles
US20020160470A1 (en) * 2000-11-13 2002-10-31 Genoptix Methods and apparatus for generating and utilizing linear moving optical gradients
US20020121443A1 (en) * 2000-11-13 2002-09-05 Genoptix Methods for the combined electrical and optical identification, characterization and/or sorting of particles
US20020123112A1 (en) * 2000-11-13 2002-09-05 Genoptix Methods for increasing detection sensitivity in optical dielectric sorting systems
AU2002241602A1 (en) * 2000-11-16 2002-06-11 Burstein Technologies, Inc. Methods and apparatus for detecting and quantifying lymphocytes with optical biodiscs
WO2002041004A2 (en) * 2000-11-16 2002-05-23 Burstein Technologies, Inc. Optical biodiscs with reflective layers
WO2002043866A2 (en) * 2000-12-01 2002-06-06 Burstein Technologies, Inc. Apparatus and methods for separating components of particulate suspension
US6760298B2 (en) * 2000-12-08 2004-07-06 Nagaoka & Co., Ltd. Multiple data layer optical discs for detecting analytes
WO2002047071A2 (en) * 2000-12-08 2002-06-13 Burstein Technologies, Inc. Multiple data layer optical discs for detecting analytes
FR2820058B1 (en) * 2001-01-29 2004-01-30 Commissariat Energie Atomique METHOD AND SYSTEM FOR MAKING A CONTINUOUS FLOW REALIZATION OF A BIOLOGICAL, CHEMICAL OR BIOCHEMICAL PROTOCOL
US20020164824A1 (en) * 2001-02-16 2002-11-07 Jianming Xiao Method and apparatus based on bundled capillaries for high throughput screening
US20020168663A1 (en) * 2001-02-27 2002-11-14 Phan Brigitte Chau Methods for DNA conjugation onto solid phase including related optical biodiscs and disc drive systems
WO2003087827A2 (en) 2001-04-11 2003-10-23 Burstein Technologies, Inc. Multi-parameter assays including analysis discs and methods relating thereto
US20030194755A1 (en) * 2001-04-27 2003-10-16 Genoptix, Inc. Early detection of apoptotic events and apoptosis using optophoretic analysis
US20040009540A1 (en) * 2001-04-27 2004-01-15 Genoptix, Inc Detection and evaluation of cancer cells using optophoretic analysis
US20030124516A1 (en) * 2001-04-27 2003-07-03 Genoptix, Inc. Method of using optical interrogation to determine a biological property of a cell or population of cells
US20040023310A1 (en) * 2001-04-27 2004-02-05 Genoptix, Inc Quantitative determination of protein kinase C activation using optophoretic analysis
WO2002087760A1 (en) * 2001-04-30 2002-11-07 Epr Labautomation Ag Method and device for storing and dosing small quantities of liquid
US20020171838A1 (en) * 2001-05-16 2002-11-21 Pal Andrew Attila Variable sampling control for rendering pixelization of analysis results in a bio-disc assembly and apparatus relating thereto
US20040166593A1 (en) * 2001-06-22 2004-08-26 Nolte David D. Adaptive interferometric multi-analyte high-speed biosensor
US6685885B2 (en) * 2001-06-22 2004-02-03 Purdue Research Foundation Bio-optical compact dist system
WO2003027723A2 (en) * 2001-07-24 2003-04-03 Burstein Technologies, Inc. Method and apparatus for bonded fluidic circuit for optical bio-disc
US20030032198A1 (en) * 2001-08-13 2003-02-13 Symyx Technologies, Inc. High throughput dispensing of fluids
GB0120131D0 (en) 2001-08-17 2001-10-10 Micromass Ltd Maldi target plate
US20030087309A1 (en) * 2001-08-27 2003-05-08 Shiping Chen Desktop drug screening system
US20030129665A1 (en) * 2001-08-30 2003-07-10 Selvan Gowri Pyapali Methods for qualitative and quantitative analysis of cells and related optical bio-disc systems
US20030143637A1 (en) * 2001-08-31 2003-07-31 Selvan Gowri Pyapali Capture layer assemblies for cellular assays including related optical analysis discs and methods
AU2002335715A1 (en) * 2001-09-07 2003-03-24 Burstein Technologies, Inc. Optical bio-disc systems for nuclear morphology based identification
WO2003042697A1 (en) * 2001-11-14 2003-05-22 Genospectra, Inc. Biochemical analysis system with combinatorial chemistry applications
EP1448992A4 (en) * 2001-11-20 2005-11-02 Burstein Technologies Inc Optical bio-discs and fluidic circuits for analysis of cells and methods relating thereto
WO2003055589A2 (en) * 2001-12-31 2003-07-10 Institut für Physikalische Hochtechnologie e.V. Microtiter plate for parallel micro synthesis, especially at high temperatures
WO2003060668A2 (en) * 2002-01-14 2003-07-24 Burstein Technologies, Inc. Method and apparatus for visualizing data
JP2005516336A (en) * 2002-01-28 2005-06-02 バースタイン テクノロジーズ,インコーポレイティド Method and apparatus for logical triggering
US20050221048A1 (en) * 2002-01-31 2005-10-06 James Rodney Norton Manufacturing processes for making optical analysis discs including successive patterning operations and optical discs thereby manufactured
US20050023765A1 (en) * 2002-01-31 2005-02-03 Coombs James Howard Bio-safety features for optical analysis disc and disc system including same
US7251210B2 (en) * 2002-01-31 2007-07-31 Burstein Technologies, Inc. Method for triggering through disc grooves and related optical analysis discs and system
WO2003064996A2 (en) * 2002-01-31 2003-08-07 Burstein Technologies, Inc. Bio-safe dispenser and optical analysis disc assembly
US20040241381A1 (en) * 2002-01-31 2004-12-02 Chen Yihfar Microfluidic structures with circumferential grooves for bonding adhesives and related optical analysis discs
FR2835615B1 (en) * 2002-02-07 2004-09-17 Antonios Vekris DEVICE FOR ANALYZING A MOLECULE BY MEANS OF A FLUID COMING INTO CONTACT WITH A SUPPORT CARRYING A MOLECULE
FR2835614A1 (en) * 2002-02-07 2003-08-08 Antonios Vekris Device for analyzing molecules, useful for nucleic acid or protein interaction assays, comprises fluid-containing chamber in which binding carrier is rotated
US6764818B2 (en) 2002-02-25 2004-07-20 Diversa Corporation Device for effecting heat transfer with a solution held in a through-hole well of a holding tray
US6918738B2 (en) 2002-03-11 2005-07-19 Diversa Corporation Stackable sample holding plate with robot removable lid
US6798520B2 (en) 2002-03-22 2004-09-28 Diversa Corporation Method for intensifying the optical detection of samples that are held in solution in the through-hole wells of a holding tray
US20030211461A1 (en) * 2002-05-01 2003-11-13 Genoptix, Inc Optophoretic detection of durgs exhibiting inhibitory effect on Bcr-Abl positive tumor cells
US20040033539A1 (en) * 2002-05-01 2004-02-19 Genoptix, Inc Method of using optical interrogation to determine a biological property of a cell or population of cells
US20030208936A1 (en) * 2002-05-09 2003-11-13 Lee Charles Hee Method for manufacturing embroidery decorated cards and envelopes
US8277753B2 (en) * 2002-08-23 2012-10-02 Life Technologies Corporation Microfluidic transfer pin
US6911132B2 (en) 2002-09-24 2005-06-28 Duke University Apparatus for manipulating droplets by electrowetting-based techniques
US7329545B2 (en) * 2002-09-24 2008-02-12 Duke University Methods for sampling a liquid flow
US20040121307A1 (en) * 2002-12-19 2004-06-24 Genoptix, Inc Early detection of cellular differentiation using optophoresis
US20040121474A1 (en) * 2002-12-19 2004-06-24 Genoptix, Inc Detection and evaluation of chemically-mediated and ligand-mediated t-cell activation using optophoretic analysis
WO2004074818A2 (en) 2002-12-20 2004-09-02 Biotrove, Inc. Assay apparatus and method using microfluidic arrays
US20060094108A1 (en) * 2002-12-20 2006-05-04 Karl Yoder Thermal cycler for microfluidic array assays
US20040129676A1 (en) * 2003-01-07 2004-07-08 Tan Roy H. Apparatus for transfer of an array of liquids and methods for manufacturing same
WO2004106925A2 (en) * 2003-03-03 2004-12-09 Nagaoka & Co.Ltd. Methods and apparatus for use in detection and quantitation of various cell types and use of optical bio-disc for performing same
US20050037484A1 (en) * 2003-04-23 2005-02-17 Norbert Staimer Optical bio-discs including spiral fluidic circuits for performing assays
WO2004113871A2 (en) * 2003-06-19 2004-12-29 Nagaoka & Co., Ltd. Fluidic circuits for sample preparation including bio-discs and methods relating thereto
US7390464B2 (en) * 2003-06-19 2008-06-24 Burstein Technologies, Inc. Fluidic circuits for sample preparation including bio-discs and methods relating thereto
WO2005001429A2 (en) * 2003-06-27 2005-01-06 Nagaoka & Co., Ltd. Fluidic circuits, methods and apparatus for use of whole blood samples in colorimetric assays
US20070274863A1 (en) * 2003-07-25 2007-11-29 Horacio Kido Fluidic circuits for sample preparation including bio-discs and methods relating thereto
CA2536360C (en) * 2003-08-28 2013-08-06 Celula, Inc. Methods and apparatus for sorting cells using an optical switch in a microfluidic channel network
US7063216B2 (en) 2003-09-04 2006-06-20 Millipore Corporation Underdrain useful in the construction of a filtration device
US9492820B2 (en) 2003-09-19 2016-11-15 Applied Biosystems, Llc High density plate filler
US7998435B2 (en) * 2003-09-19 2011-08-16 Life Technologies Corporation High density plate filler
US20060272738A1 (en) * 2003-09-19 2006-12-07 Gary Lim High density plate filler
US20050280811A1 (en) * 2003-09-19 2005-12-22 Donald Sandell Grooved high density plate
US20050220675A1 (en) * 2003-09-19 2005-10-06 Reed Mark T High density plate filler
US20050226782A1 (en) * 2003-09-19 2005-10-13 Reed Mark T High density plate filler
US20050232821A1 (en) * 2003-09-19 2005-10-20 Carrillo Albert L High density plate filler
US8277760B2 (en) * 2003-09-19 2012-10-02 Applied Biosystems, Llc High density plate filler
US20060233673A1 (en) * 2003-09-19 2006-10-19 Beard Nigel P High density plate filler
US20060233671A1 (en) * 2003-09-19 2006-10-19 Beard Nigel P High density plate filler
US7695688B2 (en) * 2003-09-19 2010-04-13 Applied Biosystems, Llc High density plate filler
US20050225751A1 (en) * 2003-09-19 2005-10-13 Donald Sandell Two-piece high density plate
US7407630B2 (en) * 2003-09-19 2008-08-05 Applera Corporation High density plate filler
US20070017870A1 (en) * 2003-09-30 2007-01-25 Belov Yuri P Multicapillary device for sample preparation
US7166212B2 (en) * 2003-09-30 2007-01-23 Chromba, Inc. Multicapillary column for chromatography and sample preparation
US8753588B2 (en) 2003-10-15 2014-06-17 Emd Millipore Corporation Support and stand-off ribs for underdrain for multi-well device
US20050264805A1 (en) * 2004-02-09 2005-12-01 Blueshift Biotechnologies, Inc. Methods and apparatus for scanning small sample volumes
AU2005222618A1 (en) 2004-03-12 2005-09-29 Biotrove, Inc. Nanoliter array loading
EP1761331A2 (en) * 2004-06-07 2007-03-14 Bioprocessors Corporation Control of reactor environmental conditions
US20060105453A1 (en) * 2004-09-09 2006-05-18 Brenan Colin J Coating process for microfluidic sample arrays
DE102004041941B4 (en) * 2004-08-30 2007-01-11 P.A.L.M. Microlaser Technologies Ag Method for obtaining biological objects with a recording unit
US7608042B2 (en) * 2004-09-29 2009-10-27 Intellidx, Inc. Blood monitoring system
US20060229531A1 (en) * 2005-02-01 2006-10-12 Daniel Goldberger Blood monitoring system
US20070191716A1 (en) * 2004-09-29 2007-08-16 Daniel Goldberger Blood monitoring system
TW200632314A (en) * 2004-12-07 2006-09-16 Honeywell Analytics Ag Gas detection method and system
PL1859330T3 (en) 2005-01-28 2013-01-31 Univ Duke Apparatuses and methods for manipulating droplets on a printed circuit board
US7910356B2 (en) * 2005-02-01 2011-03-22 Purdue Research Foundation Multiplexed biological analyzer planar array apparatus and methods
US7405831B2 (en) * 2005-02-01 2008-07-29 Purdue Research Foundation Laser scanning interferometric surface metrology
US20070023643A1 (en) * 2005-02-01 2007-02-01 Nolte David D Differentially encoded biological analyzer planar array apparatus and methods
JP4979941B2 (en) * 2005-03-30 2012-07-18 Hoya株式会社 Manufacturing method of glass substrate for mask blanks, manufacturing method of mask blanks
JP5011650B2 (en) * 2005-04-01 2012-08-29 パナソニック株式会社 Light detection method
US8357616B2 (en) * 2005-04-14 2013-01-22 President And Fellows Of Harvard College Adjustable solubility in sacrificial layers for microfabrication
US20070047388A1 (en) * 2005-08-25 2007-03-01 Rockwell Scientific Licensing, Llc Fluidic mixing structure, method for fabricating same, and mixing method
WO2007054220A1 (en) * 2005-11-09 2007-05-18 Christian Schmidt Methods and devices for surface modification of micro-structured substrates
US20080200838A1 (en) * 2005-11-28 2008-08-21 Daniel Goldberger Wearable, programmable automated blood testing system
JP4861042B2 (en) * 2006-04-17 2012-01-25 株式会社日立ハイテクマニファクチャ&サービス Spectrophotometer
US20070259366A1 (en) * 2006-05-03 2007-11-08 Greg Lawrence Direct printing of patterned hydrophobic wells
US8092385B2 (en) * 2006-05-23 2012-01-10 Intellidx, Inc. Fluid access interface
WO2008046112A1 (en) * 2006-10-13 2008-04-17 Eliseev Alexey V Methods and microarrays compatible with dual functionality optical drives
US7522282B2 (en) * 2006-11-30 2009-04-21 Purdue Research Foundation Molecular interferometric imaging process and apparatus
US20080144899A1 (en) * 2006-11-30 2008-06-19 Manoj Varma Process for extracting periodic features from images by template matching
US20080230605A1 (en) * 2006-11-30 2008-09-25 Brian Weichel Process and apparatus for maintaining data integrity
US7659968B2 (en) * 2007-01-19 2010-02-09 Purdue Research Foundation System with extended range of molecular sensing through integrated multi-modal data acquisition
WO2008118934A1 (en) * 2007-03-26 2008-10-02 Purdue Research Foundation Method and apparatus for conjugate quadrature interferometric detection of an immunoassay
WO2009021233A2 (en) 2007-08-09 2009-02-12 Advanced Liquid Logic, Inc. Pcb droplet actuator fabrication
JP5295733B2 (en) 2007-11-30 2013-09-18 キヤノン株式会社 Living body holding method, living body testing method, living body growth method, living body holding sheet, and living body processing apparatus
JP4518157B2 (en) * 2008-01-31 2010-08-04 カシオ計算機株式会社 Imaging apparatus and program thereof
WO2009121034A2 (en) * 2008-03-28 2009-10-01 Pelican Group Holdings, Inc. Multicapillary sample preparation devices and methods for processing analytes
CN102026723B (en) * 2008-03-31 2015-03-25 凯杰博登湖有限公司 Sample holder and method of using the same
US8704158B2 (en) 2008-04-17 2014-04-22 Qiagen Lake Constance Gmbh Fluorescence standard, and the use thereof
JP5283113B2 (en) * 2008-10-03 2013-09-04 独立行政法人産業技術総合研究所 Method and apparatus for measuring volume of microdroplet
US8753290B2 (en) * 2009-03-27 2014-06-17 Intellectual Inspiration, Llc Fluid transfer system and method
JP5550262B2 (en) * 2009-05-29 2014-07-16 キヤノン株式会社 Sample observation system and sample observation method
KR20120123066A (en) * 2009-12-31 2012-11-07 바스프 에스이 Tampering detector and method
GB2480596A (en) * 2010-01-29 2011-11-30 Kenneth John Cunningham Disposable light scattering cuvette for liquid sample held by surface tension
CN103282123B (en) 2010-12-30 2015-05-06 艾博特健康公司 Biologic fluid analysis cartridge with sample handling portion and analysis chamber portion
WO2013049659A2 (en) * 2011-09-30 2013-04-04 Life Technologies Corporation Systems and methods for biological analysis
JP5819986B2 (en) * 2011-12-19 2015-11-24 ヤマハ発動機株式会社 Object sorting apparatus and object sorting method
KR101309129B1 (en) 2012-03-15 2013-09-16 주식회사 메카시스 Sample handler for analyzing a small quantity of sample, analyzing apparatus using the sample handler, and analyzing method
JP2015511015A (en) * 2012-03-16 2015-04-13 ライフ テクノロジーズ コーポレーション System and method for containing biological samples
EP3984643A1 (en) 2012-03-16 2022-04-20 Life Technologies Corporation Systems and methods for assessing of biological samples
JP5483134B2 (en) * 2012-10-09 2014-05-07 独立行政法人産業技術総合研究所 Method and apparatus for measuring volume of microdroplet
KR101387576B1 (en) 2012-11-08 2014-04-23 주식회사 신코 Micro-volume sample holder
US8711351B1 (en) 2013-01-29 2014-04-29 Hewlett-Packard Development Company, L.P. Scattering spectroscopy employing hotspot-aligned nanopores
US11085067B2 (en) 2013-06-10 2021-08-10 President And Fellows Of Harvard College Early developmental genomic assay for characterizing pluripotent stem cell utility and safety
EP3142792A2 (en) * 2014-05-14 2017-03-22 University of Limerick Microfluidic devices that include channels that are slidable relative to each other and methods of use thereof
US10376888B2 (en) 2014-07-03 2019-08-13 Centrillion Technology Holdings Corporation Device for storage and dispensing of reagents
EP3043172B1 (en) * 2015-01-06 2018-06-27 Rolls-Royce plc Method and apparatus for testing of engine components
US10307724B2 (en) 2015-07-02 2019-06-04 Centrillion Technology Holdings Corporation Systems and methods to dispense and mix reagents
US11085039B2 (en) 2016-12-12 2021-08-10 xCella Biosciences, Inc. Methods and systems for screening using microcapillary arrays
JP7356351B2 (en) 2016-12-12 2023-10-04 エクセラ・バイオサイエンシーズ・インコーポレイテッド Methods and systems for screening using microcapillary arrays
CN110382439A (en) 2016-12-30 2019-10-25 埃克切拉生物科学公司 Multistage sample retrieval system
EP3837344A4 (en) * 2018-08-17 2022-01-26 Sierra Biosystems, Inc. Row-independent oligonucleotide synthesis
WO2020043906A1 (en) * 2018-08-31 2020-03-05 Oerlikon Am Gmbh Alloy development instrument for additive manufacturing (am)
EP3890876A4 (en) 2018-12-06 2022-06-08 Xcella Biosciences, Inc. Lateral loading of microcapillary arrays
TW202100247A (en) * 2019-01-29 2021-01-01 美商伊路米納有限公司 Flow cells
WO2021134014A1 (en) * 2019-12-27 2021-07-01 Sean Kelly Analysis of a biological sample using tape-to-tape fluidic transfer
FR3116008A1 (en) * 2020-11-06 2022-05-13 Preciphos Production of biofunctionalized filter chips

Citations (38)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2771398A (en) * 1953-09-17 1956-11-20 Thomas L Snyder Method and apparatus for counting microorganisms
US3036893A (en) * 1960-03-14 1962-05-29 Scientific Industries Automatic chemical analyzer
US3098719A (en) * 1960-06-16 1963-07-23 Technicon Instr Fluid processing system with continuous filter apparatus
US3216804A (en) * 1962-01-31 1965-11-09 Scientific Industries Automatic chemical analyzer and sample dispenser
US3260413A (en) * 1964-08-31 1966-07-12 Scientific Industries Automatic chemical analyzer
US3261668A (en) * 1962-08-14 1966-07-19 Scientific Industries Chemical analyzer tape
US3475128A (en) * 1966-04-08 1969-10-28 Bio Science Labor Fluid processing apparatus and methods
US3497320A (en) * 1966-12-15 1970-02-24 Xerox Corp Automated chemical analyzer
US3508879A (en) * 1966-12-15 1970-04-28 Xerox Corp Aliquotting device
US3526480A (en) * 1966-12-15 1970-09-01 Xerox Corp Automated chemical analyzer
US3554700A (en) * 1968-05-06 1971-01-12 Scientific Industries Method for obtaining a known volume of liquid and absorption apparatus therefor
US3607090A (en) * 1969-10-06 1971-09-21 Scientific Industries Analysis arrangment for multiple analyses of a single sample
US3607079A (en) * 1969-06-02 1971-09-21 Scientific Industries Analysis arrangement for chemical analyzing apparatus
US3620678A (en) * 1966-09-08 1971-11-16 Jean Guigan Installation for multiple and automatic analyses
US3675488A (en) * 1969-09-11 1972-07-11 Res Foundation Of The Washingt Apparatus for transport and storage of liquid specimens for radio-immunoassay for insulin
US3676080A (en) * 1967-12-15 1972-07-11 Hoffmann La Roche Device for automatically analyzing liquids
US3698822A (en) * 1969-03-19 1972-10-17 American Optical Corp Disposable cuvettes for colorimetry
US3979264A (en) * 1975-03-31 1976-09-07 Heinz Buerger Band for carrying out microbiological examinations
US4071315A (en) * 1976-06-02 1978-01-31 Guy Chateau Analytical tape medium
US4263256A (en) * 1979-11-05 1981-04-21 Coulter Electronics, Inc. Cuvettes for automatic chemical apparatus
US4327073A (en) * 1980-04-07 1982-04-27 Huang Henry V Automated method for quantitative analysis of biological fluids
US4349510A (en) * 1979-07-24 1982-09-14 Seppo Kolehmainen Method and apparatus for measurement of samples by luminescence
US4568520A (en) * 1982-06-05 1986-02-04 Ismatec Sa Apparatus for the automated production of a series of samples for the analysis of mixtures
US4682890A (en) * 1985-05-31 1987-07-28 Health Research, Incorporated Microsample holder and carrier therefor
US4685880A (en) * 1985-06-18 1987-08-11 American Hospital Supply Corporation Cuvette belts and manufacture of same
US4853059A (en) * 1986-12-24 1989-08-01 Baxter International Inc. Apparatus and process for manufacturing cuvetter belts
US4863693A (en) * 1984-08-21 1989-09-05 E. I. Du Pont De Nemours And Company Analysis instrument having a blow molded reaction chamber
US4883642A (en) * 1984-06-05 1989-11-28 Universite Paris-Nord Means to automatically hold, process, store and analyze biological samples
US5009850A (en) * 1988-05-05 1991-04-23 Smiths Industries Medical Systems, Inc. Blood containment device
US5077010A (en) * 1987-07-15 1991-12-31 Fuji Photo Film Co., Ltd. Long-test-film cassette for biochemical analysis, and system for loading the same
US5213766A (en) * 1991-04-30 1993-05-25 Apogee Designs, Ltd. Liquid collecting apparatus for sample testing
US5290705A (en) * 1992-01-13 1994-03-01 R. E. Davis Chemical Corporation Speciman support for optical analysis
US5508200A (en) * 1992-10-19 1996-04-16 Tiffany; Thomas Method and apparatus for conducting multiple chemical assays
US5985594A (en) * 1995-11-14 1999-11-16 Idexx Laboratories, Inc. Method for quantification of biological material in a sample
US6027873A (en) * 1999-03-19 2000-02-22 Genencor International, Inc. Multi-through hole testing plate for high throughput screening
US6387331B1 (en) * 1998-01-12 2002-05-14 Massachusetts Institute Of Technology Method and apparatus for performing microassays
US6565813B1 (en) * 1998-02-04 2003-05-20 Merck & Co., Inc. Virtual wells for use in high throughput screening assays
US6878345B1 (en) * 1997-12-08 2005-04-12 Thomas W. Astle Ultra high throughput bioassay screening system

Family Cites Families (126)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3627431A (en) * 1969-12-22 1971-12-14 John Victor Komarniski Densitometer
US3645696A (en) * 1970-04-30 1972-02-29 Cities Service Oil Co Method for stabilizing chromogenic test reagent for aldehyde
JPS5518906B1 (en) * 1971-02-18 1980-05-22
US3768974A (en) * 1971-03-22 1973-10-30 Sterilizer Control Royalties Disposable colorimetric indicator device for measuring the concentration of chlorine in water
US3666421A (en) * 1971-04-05 1972-05-30 Organon Diagnostic test slide
GB1320426A (en) 1971-07-06 1973-06-13 Pfizer Multiple solution testing device
US3950133A (en) * 1971-10-20 1976-04-13 Mallinckrodt, Inc. Reagent formulations for assaying biological specimens and methods of preparing and using same
US3994594A (en) * 1975-08-27 1976-11-30 Technicon Instruments Corporation Cuvette and method of use
SE399768B (en) * 1975-09-29 1978-02-27 Lilja Jan E CYVETT FOR SAMPLING, MIXING OF, THE SAMPLE WITH A REAGENTS AND DIRECT PERFORMANCE OF, SPECIAL OPTICAL, ANALYSIS OF THE SAMPLE MIXED WITH THE REAGENTS
US4111754A (en) 1976-11-29 1978-09-05 Hydow Park Immunological testing devices and methods
US4407943A (en) * 1976-12-16 1983-10-04 Millipore Corporation Immobilized antibody or antigen for immunoassay
US4273877A (en) * 1978-06-13 1981-06-16 National Research Development Corporation Spiral plating apparatus
US4234316A (en) * 1979-04-02 1980-11-18 Fmc Corporation Device for delivering measured quantities of reagents into assay medium
US4500707A (en) * 1980-02-29 1985-02-19 University Patents, Inc. Nucleosides useful in the preparation of polynucleotides
US4387164A (en) * 1980-11-05 1983-06-07 Fmc Corporation Method and apparatus for chemical analysis using reactive reagents dispersed in soluble film
US4437109A (en) * 1980-11-07 1984-03-13 General Electric Company Silicon-on-sapphire body with conductive paths therethrough
US4394712A (en) * 1981-03-18 1983-07-19 General Electric Company Alignment-enhancing feed-through conductors for stackable silicon-on-sapphire wafers
US4973679A (en) * 1981-03-27 1990-11-27 University Patents, Inc. Process for oligonucleo tide synthesis using phosphormidite intermediates
US4415732A (en) * 1981-03-27 1983-11-15 University Patents, Inc. Phosphoramidite compounds and processes
JPS57175957A (en) 1981-04-24 1982-10-29 Chugai Pharmaceut Co Ltd Measuring method and device for antigen- antibody reaction
US4527183A (en) * 1981-07-10 1985-07-02 General Electric Company Drilled, diffused radiation detector
US4473737A (en) * 1981-09-28 1984-09-25 General Electric Company Reverse laser drilling
US4562045A (en) * 1981-10-07 1985-12-31 Murata Manufacturing Co. Carrier for holding analytical samples
US5310674A (en) 1982-05-10 1994-05-10 Bar-Ilan University Apertured cell carrier
US4861448A (en) * 1982-11-18 1989-08-29 The Trustees Of Columbia University In The City Of New York Electrophoretic methods employing gel inserts
US4761378A (en) * 1983-03-04 1988-08-02 American Home Products Corp. (Del.) Microbiological testing apparatus
US4493815A (en) 1983-07-28 1985-01-15 Bio-Rad Laboratories, Inc. Supporting and filtering biochemical test plate assembly
US4547836A (en) * 1984-02-01 1985-10-15 General Electric Company Insulating glass body with electrical feedthroughs and method of preparation
US4562871A (en) * 1984-03-16 1986-01-07 Astle Thomas W Rehydrator
US4586546A (en) * 1984-10-23 1986-05-06 Cetus Corporation Liquid handling device and method
US4682891A (en) * 1985-05-31 1987-07-28 Health Research, Incorporated Microcircle system
US5047215A (en) 1985-06-18 1991-09-10 Polyfiltronics, Inc. Multiwell test plate
JPS62144048A (en) * 1985-12-18 1987-06-27 Olympus Optical Co Ltd Developer concentration measuring apparatus
US4728591A (en) * 1986-03-07 1988-03-01 Trustees Of Boston University Self-assembled nanometer lithographic masks and templates and method for parallel fabrication of nanometer scale multi-device structures
US5153319A (en) * 1986-03-31 1992-10-06 University Patents, Inc. Process for preparing polynucleotides
JPS63241359A (en) * 1986-10-01 1988-10-06 Olympus Optical Co Ltd Micromodule solid phase method of reagent
US5000921A (en) 1986-10-24 1991-03-19 Hanaway Richard W Multiple pipette samples
US4777143A (en) * 1986-12-12 1988-10-11 Litmus Concepts Inc. Method of detecting carboxylic acids in a specimen
US4834946A (en) 1987-02-05 1989-05-30 Levin Andrew E Apparatus for blot screening numerous, small volume, antibody solutions
US5077085A (en) 1987-03-06 1991-12-31 Schnur Joel M High resolution metal patterning of ultra-thin films on solid substrates
US4806316A (en) * 1987-03-17 1989-02-21 Becton, Dickinson And Company Disposable device for use in chemical, immunochemical and microorganism analysis
US4828386A (en) 1987-06-19 1989-05-09 Pall Corporation Multiwell plates containing membrane inserts
US4990459A (en) * 1988-04-25 1991-02-05 Kabushiki Kaisha Toshiba Impurity measuring method
US4892409A (en) * 1988-07-14 1990-01-09 Smith Harry F Photometric apparatus for multiwell plates having a positionable lens assembly
WO1990002326A1 (en) * 1988-08-23 1990-03-08 Bio-Mediq (Australia) Pty. Ltd. Optical fluid analysis imaging and positioning
US6147198A (en) * 1988-09-15 2000-11-14 New York University Methods and compositions for the manipulation and characterization of individual nucleic acid molecules
US5108704A (en) 1988-09-16 1992-04-28 W. R. Grace & Co.-Conn. Microfiltration apparatus with radially spaced nozzles
FR2637687B1 (en) * 1988-10-11 1991-01-11 Inst Textile De France SINGLE USE DEVICE FOR BIOLOGICAL TESTS
EP0391674B1 (en) * 1989-04-05 1996-03-20 New York University A method for characterising particles
US5744101A (en) * 1989-06-07 1998-04-28 Affymax Technologies N.V. Photolabile nucleoside protecting groups
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
US5183761A (en) * 1989-07-21 1993-02-02 Freeman Mary J Method of making calibration solution for verifying calibration and linearity of vertical photometers
US5262128A (en) 1989-10-23 1993-11-16 The United States Of America As Represented By The Department Of Health And Human Services Array-type multiple cell injector
US5229163A (en) * 1989-12-21 1993-07-20 Hoffmann-La Roche Inc. Process for preparing a microtiter tray for immunometric determinations
US5621094A (en) * 1990-05-14 1997-04-15 Quadrant Holdings Cambridge Limited Method of preserving agarose gel structure during dehydration by adding a non-reducing glycoside of a straight-chain sugar alcohol
JPH0678978B2 (en) * 1990-05-25 1994-10-05 スズキ株式会社 Aggregation pattern detector
JPH04169839A (en) * 1990-11-01 1992-06-17 Mitsubishi Electric Corp Alcohol-content detecting apparatus
JPH04250874A (en) * 1991-01-09 1992-09-07 Canon Inc Liquid introducing device
US5182082A (en) * 1991-01-23 1993-01-26 Becton, Dickinson And Company Multiple aliquot device for distributing a liquid solution into a well
US5955377A (en) * 1991-02-11 1999-09-21 Biostar, Inc. Methods and kits for the amplification of thin film based assays
US6004744A (en) * 1991-03-05 1999-12-21 Molecular Tool, Inc. Method for determining nucleotide identity through extension of immobilized primer
US5284753A (en) 1991-03-20 1994-02-08 Neuro Probe, Inc. Multiple-site chemotactic test apparatus and method
US5210021A (en) 1991-03-20 1993-05-11 Neuro Probe, Inc. Multiple-site chemotactic test apparatus and method
US5632957A (en) * 1993-11-01 1997-05-27 Nanogen Molecular biological diagnostic systems including electrodes
US5605662A (en) * 1993-11-01 1997-02-25 Nanogen, Inc. Active programmable electronic devices for molecular biological analysis and diagnostics
US5888723A (en) * 1992-02-18 1999-03-30 Johnson & Johnson Clinical Diagnostics, Inc. Method for nucleic acid amplification and detection using adhered probes
US5374525A (en) * 1992-09-30 1994-12-20 University Of Utah Research Foundation Methods to determine predisposition to hypertension and association of variant angiotensinogen gene and hypertension
US5455934A (en) * 1993-03-23 1995-10-03 Eclipse Technologies, Inc. Fault tolerant hard disk array controller
WO1995001559A2 (en) * 1993-07-02 1995-01-12 Evotec Biosystems Gmbh Sample holder and its use
US5519218A (en) * 1993-08-04 1996-05-21 Chang; On Kok Sample holder for spectroscopy
WO1995011755A1 (en) * 1993-10-28 1995-05-04 Houston Advanced Research Center Microfabricated, flowthrough porous apparatus for discrete detection of binding reactions
US5553616A (en) * 1993-11-30 1996-09-10 Florida Institute Of Technology Determination of concentrations of biological substances using raman spectroscopy and artificial neural network discriminator
US5578832A (en) * 1994-09-02 1996-11-26 Affymetrix, Inc. Method and apparatus for imaging a sample on a device
FR2716263B1 (en) * 1994-02-11 1997-01-17 Pasteur Institut Method for aligning macromolecules by passing a meniscus and applications in a method for highlighting, separating and / or assaying a macromolecule in a sample.
US5453252A (en) * 1994-02-25 1995-09-26 Truett; William L. Screen cell for spectroscopy
US5807522A (en) * 1994-06-17 1998-09-15 The Board Of Trustees Of The Leland Stanford Junior University Methods for fabricating microarrays of biological samples
US5515167A (en) * 1994-09-13 1996-05-07 Hughes Aircraft Company Transparent optical chuck incorporating optical monitoring
US6121048A (en) * 1994-10-18 2000-09-19 Zaffaroni; Alejandro C. Method of conducting a plurality of reactions
DE19509094A1 (en) * 1995-03-16 1996-09-26 Boehringer Mannheim Gmbh Quantitative transmission spectroscopy using sample carriers with networks
JPH08254536A (en) * 1995-03-17 1996-10-01 Toshiba Corp Automatic chemical analyzer
US5560811A (en) 1995-03-21 1996-10-01 Seurat Analytical Systems Incorporated Capillary electrophoresis apparatus and method
US5609828A (en) * 1995-05-31 1997-03-11 bio M erieux Vitek, Inc. Sample card
US5545531A (en) * 1995-06-07 1996-08-13 Affymax Technologies N.V. Methods for making a device for concurrently processing multiple biological chip assays
CA2179364C (en) 1995-06-27 1999-09-28 Klaus W. Berndt Method and apparatus for detecting microorganisms
US5773238A (en) * 1995-07-07 1998-06-30 Shukla; Ashok K. Droplet chemical reaction chamber
EP2275821A1 (en) * 1995-07-31 2011-01-19 Precision System Science Co., Ltd Container
GB9521775D0 (en) 1995-10-24 1996-01-03 Pa Consulting Services Microwell plates
US5763263A (en) * 1995-11-27 1998-06-09 Dehlinger; Peter J. Method and apparatus for producing position addressable combinatorial libraries
USH1919H (en) * 1995-12-01 2000-11-07 E. I. Du Pont De Nemours And Company Agricultural product microscreen method and apparatus
CA2192262C (en) * 1995-12-08 2011-03-15 Yoshihide Hayashizaki Method for purification and transfer to separation/detection systems of dna sequencing samples and plates used therefor
US6660233B1 (en) * 1996-01-16 2003-12-09 Beckman Coulter, Inc. Analytical biochemistry system with robotically carried bioarray
US5849598A (en) * 1996-03-15 1998-12-15 Washington University Method for transferring micro quantities of liquid samples to discrete locations
GB9606367D0 (en) * 1996-03-26 1996-06-05 United Utilities Plc Optical instrument
US6001586A (en) * 1996-03-29 1999-12-14 Genencor International, Inc. Compartmentalization method for screening microorganisms
US5788814A (en) * 1996-04-09 1998-08-04 David Sarnoff Research Center Chucks and methods for positioning multiple objects on a substrate
US5840256A (en) * 1996-04-09 1998-11-24 David Sarnoff Research Center Inc. Plate for reaction system
US6103479A (en) * 1996-05-30 2000-08-15 Cellomics, Inc. Miniaturized cell array methods and apparatus for cell-based screening
DE19628178C1 (en) * 1996-07-12 1997-09-18 Bruker Franzen Analytik Gmbh Loading matrix-assisted laser desorption-ionisation sample plate for mass spectrometric analysis
IL128539A (en) * 1996-08-16 2004-01-04 Imaging Res Inc Digital imaging system for assays in well plates, gels and blots
US5795748A (en) * 1996-09-26 1998-08-18 Becton Dickinson And Company DNA microwell device and method
US5854684A (en) * 1996-09-26 1998-12-29 Sarnoff Corporation Massively parallel detection
US6024925A (en) * 1997-01-23 2000-02-15 Sequenom, Inc. Systems and methods for preparing low volume analyte array elements
US6060240A (en) * 1996-12-13 2000-05-09 Arcaris, Inc. Methods for measuring relative amounts of nucleic acids in a complex mixture and retrieval of specific sequences therefrom
US5944652A (en) * 1996-12-27 1999-08-31 City Of Hope Method for breeding chickens
SE509274C2 (en) * 1997-03-11 1999-01-11 Gematron Medical Ab Spectrophotometric analysis cuvette
US5958345A (en) * 1997-03-14 1999-09-28 Moxtek, Inc. Thin film sample support
EP0973863B1 (en) * 1997-04-09 2005-05-25 Minnesota Mining And Manufacturing Company Method and devices for partitioning biological sample liquids into microvolumes
US6143496A (en) * 1997-04-17 2000-11-07 Cytonix Corporation Method of sampling, amplifying and quantifying segment of nucleic acid, polymerase chain reaction assembly having nanoliter-sized sample chambers, and method of filling assembly
US5985214A (en) * 1997-05-16 1999-11-16 Aurora Biosciences Corporation Systems and methods for rapidly identifying useful chemicals in liquid samples
US5910287A (en) * 1997-06-03 1999-06-08 Aurora Biosciences Corporation Low background multi-well plates with greater than 864 wells for fluorescence measurements of biological and biochemical samples
US5922604A (en) * 1997-06-05 1999-07-13 Gene Tec Corporation Thin reaction chambers for containing and handling liquid microvolumes
US6090251A (en) * 1997-06-06 2000-07-18 Caliper Technologies, Inc. Microfabricated structures for facilitating fluid introduction into microfluidic devices
US6071748A (en) * 1997-07-16 2000-06-06 Ljl Biosystems, Inc. Light detection device
US6045753A (en) * 1997-07-29 2000-04-04 Sarnoff Corporation Deposited reagents for chemical processes
US5804437A (en) * 1997-08-19 1998-09-08 Biomerieux Vitek, Inc. Locking structure for securing a fluid transfer tube
US6309600B1 (en) * 1997-08-28 2001-10-30 Biotrove, Inc. Apparatus for droplet microchemistry
JP2001521169A (en) * 1997-10-24 2001-11-06 ノースイースタン・ユニバーシティ Multi-channel micro-scale system for high-throughput coordinated separation with extensive collection and analysis
US6893877B2 (en) * 1998-01-12 2005-05-17 Massachusetts Institute Of Technology Methods for screening substances in a microwell array
JP3445501B2 (en) * 1998-09-04 2003-09-08 日立ソフトウエアエンジニアリング株式会社 Manufacturing method of sheet-like member laminate and sheet-like probe holder
US6689323B2 (en) * 1998-10-30 2004-02-10 Agilent Technologies Method and apparatus for liquid transfer
US6309828B1 (en) * 1998-11-18 2001-10-30 Agilent Technologies, Inc. Method and apparatus for fabricating replicate arrays of nucleic acid molecules
US6296702B1 (en) * 1999-03-15 2001-10-02 Pe Corporation (Ny) Apparatus and method for spotting a substrate
WO2000056456A1 (en) * 1999-03-19 2000-09-28 Genencor International, Inc. Multi-through hole testing plate for high throughput screening
US6713309B1 (en) * 1999-07-30 2004-03-30 Large Scale Proteomics Corporation Microarrays and their manufacture
CA2400644C (en) * 2000-02-18 2009-07-14 Board Of Trustees Of The Leland Stanford Junior University Apparatus and methods for parallel processing of micro-volume liquid reactions
WO2002030561A2 (en) * 2000-10-10 2002-04-18 Biotrove, Inc. Apparatus for assay, synthesis and storage, and methods of manufacture, use, and manipulation thereof

Patent Citations (38)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2771398A (en) * 1953-09-17 1956-11-20 Thomas L Snyder Method and apparatus for counting microorganisms
US3036893A (en) * 1960-03-14 1962-05-29 Scientific Industries Automatic chemical analyzer
US3098719A (en) * 1960-06-16 1963-07-23 Technicon Instr Fluid processing system with continuous filter apparatus
US3216804A (en) * 1962-01-31 1965-11-09 Scientific Industries Automatic chemical analyzer and sample dispenser
US3261668A (en) * 1962-08-14 1966-07-19 Scientific Industries Chemical analyzer tape
US3260413A (en) * 1964-08-31 1966-07-12 Scientific Industries Automatic chemical analyzer
US3475128A (en) * 1966-04-08 1969-10-28 Bio Science Labor Fluid processing apparatus and methods
US3620678A (en) * 1966-09-08 1971-11-16 Jean Guigan Installation for multiple and automatic analyses
US3508879A (en) * 1966-12-15 1970-04-28 Xerox Corp Aliquotting device
US3526480A (en) * 1966-12-15 1970-09-01 Xerox Corp Automated chemical analyzer
US3497320A (en) * 1966-12-15 1970-02-24 Xerox Corp Automated chemical analyzer
US3676080A (en) * 1967-12-15 1972-07-11 Hoffmann La Roche Device for automatically analyzing liquids
US3554700A (en) * 1968-05-06 1971-01-12 Scientific Industries Method for obtaining a known volume of liquid and absorption apparatus therefor
US3698822A (en) * 1969-03-19 1972-10-17 American Optical Corp Disposable cuvettes for colorimetry
US3607079A (en) * 1969-06-02 1971-09-21 Scientific Industries Analysis arrangement for chemical analyzing apparatus
US3675488A (en) * 1969-09-11 1972-07-11 Res Foundation Of The Washingt Apparatus for transport and storage of liquid specimens for radio-immunoassay for insulin
US3607090A (en) * 1969-10-06 1971-09-21 Scientific Industries Analysis arrangment for multiple analyses of a single sample
US3979264A (en) * 1975-03-31 1976-09-07 Heinz Buerger Band for carrying out microbiological examinations
US4071315A (en) * 1976-06-02 1978-01-31 Guy Chateau Analytical tape medium
US4349510A (en) * 1979-07-24 1982-09-14 Seppo Kolehmainen Method and apparatus for measurement of samples by luminescence
US4263256A (en) * 1979-11-05 1981-04-21 Coulter Electronics, Inc. Cuvettes for automatic chemical apparatus
US4327073A (en) * 1980-04-07 1982-04-27 Huang Henry V Automated method for quantitative analysis of biological fluids
US4568520A (en) * 1982-06-05 1986-02-04 Ismatec Sa Apparatus for the automated production of a series of samples for the analysis of mixtures
US4883642A (en) * 1984-06-05 1989-11-28 Universite Paris-Nord Means to automatically hold, process, store and analyze biological samples
US4863693A (en) * 1984-08-21 1989-09-05 E. I. Du Pont De Nemours And Company Analysis instrument having a blow molded reaction chamber
US4682890A (en) * 1985-05-31 1987-07-28 Health Research, Incorporated Microsample holder and carrier therefor
US4685880A (en) * 1985-06-18 1987-08-11 American Hospital Supply Corporation Cuvette belts and manufacture of same
US4853059A (en) * 1986-12-24 1989-08-01 Baxter International Inc. Apparatus and process for manufacturing cuvetter belts
US5077010A (en) * 1987-07-15 1991-12-31 Fuji Photo Film Co., Ltd. Long-test-film cassette for biochemical analysis, and system for loading the same
US5009850A (en) * 1988-05-05 1991-04-23 Smiths Industries Medical Systems, Inc. Blood containment device
US5213766A (en) * 1991-04-30 1993-05-25 Apogee Designs, Ltd. Liquid collecting apparatus for sample testing
US5290705A (en) * 1992-01-13 1994-03-01 R. E. Davis Chemical Corporation Speciman support for optical analysis
US5508200A (en) * 1992-10-19 1996-04-16 Tiffany; Thomas Method and apparatus for conducting multiple chemical assays
US5985594A (en) * 1995-11-14 1999-11-16 Idexx Laboratories, Inc. Method for quantification of biological material in a sample
US6878345B1 (en) * 1997-12-08 2005-04-12 Thomas W. Astle Ultra high throughput bioassay screening system
US6387331B1 (en) * 1998-01-12 2002-05-14 Massachusetts Institute Of Technology Method and apparatus for performing microassays
US6565813B1 (en) * 1998-02-04 2003-05-20 Merck & Co., Inc. Virtual wells for use in high throughput screening assays
US6027873A (en) * 1999-03-19 2000-02-22 Genencor International, Inc. Multi-through hole testing plate for high throughput screening

Also Published As

Publication number Publication date
EP1051259A1 (en) 2000-11-15
JP4448166B2 (en) 2010-04-07
AU2102699A (en) 1999-07-26
US20170028376A9 (en) 2017-02-02
JP4271371B2 (en) 2009-06-03
US6387331B1 (en) 2002-05-14
ATE322341T1 (en) 2006-04-15
EP1714699A1 (en) 2006-10-25
US20040171166A1 (en) 2004-09-02
DE69930726D1 (en) 2006-05-18
JP2002500373A (en) 2002-01-08
EP1714699B1 (en) 2010-08-18
US6743633B1 (en) 2004-06-01
ES2350702T3 (en) 2011-01-26
US20150126412A1 (en) 2015-05-07
EP2286918B1 (en) 2014-10-01
CA2316912C (en) 2009-09-15
EP1051259B1 (en) 2006-04-05
US20110065590A1 (en) 2011-03-17
DE69942697D1 (en) 2010-09-30
JP4668334B2 (en) 2011-04-13
JP2009244271A (en) 2009-10-22
DE69930726T2 (en) 2007-01-25
ATE477850T1 (en) 2010-09-15
CA2316912A1 (en) 1999-07-15
JP2008139319A (en) 2008-06-19
WO1999034920A1 (en) 1999-07-15
EP2286918A1 (en) 2011-02-23

Similar Documents

Publication Publication Date Title
US7547556B2 (en) Methods for filing a sample array by droplet dragging
US20150298089A1 (en) Systems for Filling a Sample Array by Droplet Dragging
US6716629B2 (en) Apparatus for assay, synthesis and storage, and methods of manufacture, use, and manipulation thereof
US9968903B2 (en) Apparatus for assay, synthesis and storage, and methods of manufacture, use, and manipulation thereof
JP4387588B2 (en) Virtual wells for high-throughput screening assays

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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